Language selection

Search

Patent 2464472 Summary

Third-party information liability

Some of the information on this Web page has been provided by external sources. The Government of Canada is not responsible for the accuracy, reliability or currency of the information supplied by external sources. Users wishing to rely upon this information should consult directly with the source of the information. Content provided by external sources is not subject to official languages, privacy and accessibility requirements.

Claims and Abstract availability

Any discrepancies in the text and image of the Claims and Abstract are due to differing posting times. Text of the Claims and Abstract are posted:

  • At the time the application is open to public inspection;
  • At the time of issue of the patent (grant).
(12) Patent: (11) CA 2464472
(54) English Title: ANTIBODY TARGETING COMPOUNDS
(54) French Title: COMPOSES DE CIBLAGE D'ANTICORPS
Status: Expired
Bibliographic Data
(51) International Patent Classification (IPC):
  • C07K 16/00 (2006.01)
  • A61K 39/395 (2006.01)
  • A61K 49/00 (2006.01)
  • A61P 31/00 (2006.01)
  • C07K 16/40 (2006.01)
  • C07K 19/00 (2006.01)
  • G01N 33/53 (2006.01)
  • A61K 51/10 (2006.01)
  • A61K 47/48 (2006.01)
(72) Inventors :
  • BARBAS, CARLOS F. (United States of America)
  • RADER, CHRISTOPH (United States of America)
  • SINHA, SUBHASH C. (United States of America)
  • LERNER, RICHARD (United States of America)
(73) Owners :
  • THE SCRIPPS RESEARCH INSTITUTE (United States of America)
(71) Applicants :
  • THE SCRIPPS RESEARCH INSTITUTE (United States of America)
(74) Agent: SMART & BIGGAR LP
(74) Associate agent:
(45) Issued: 2014-01-07
(86) PCT Filing Date: 2002-10-22
(87) Open to Public Inspection: 2003-07-24
Examination requested: 2007-06-01
Availability of licence: N/A
(25) Language of filing: English

Patent Cooperation Treaty (PCT): Yes
(86) PCT Filing Number: PCT/US2002/033991
(87) International Publication Number: WO2003/059251
(85) National Entry: 2004-04-20

(30) Application Priority Data:
Application No. Country/Territory Date
60/344,614 United States of America 2001-10-22
60/412,455 United States of America 2002-09-19

Abstracts

English Abstract




The present invention provides antibody targeting compounds in which the
specificity of the antibody has been reprogrammed by covalently or
noncovalently linking a targeting agent to the combining site of an antibody.
By this approach, the covalently modified antibody takes on the binding
specificity of the targeting agent. The compound may have biological activity
provided by the targeting agent or by a separate biological agent. Various
uses of the invention compounds are provided.


French Abstract

La présente invention a trait à des composés de ciblage d'anticorps, dans lesquels la spécificité de l'anticorps a été reprogrammée par l'établissement d'une liaison covalente ou non covalente entre un agent de ciblage et le site anticorps d'un anticorps. Par cette approche, l'anticorps modifié par covalence prend en charge la spécificité de liaison de l'agent de ciblage. Le composé peut présenter une activité biologique assurée par l'agent de ciblage ou par un autre agent biologique. L'invention concerne également diverses utilisations des composés de l'invention.

Claims

Note: Claims are shown in the official language in which they were submitted.



CLAIMS:

1. An antibody targeting compound comprising at least one targeting agent
at
least 500 daltons in size covalently linked to the combining site of a
catalytic aldolase
antibody via a linker of formula X-Y-Z, wherein the X group is a linear or
branched
connecting chain of atoms comprising any of C, H, N, O, P, S, Si, F, CI, Br,
and I, or a salt
thereof, the Y group is an optional recognition group comprising one or more
ring structures,
and the Z group is a reactive group:
Image
wherein Y refers to the Y group of the linker or X group if Y is absent;
wherein the targeting agent is linked to the X group of the linker; and
whereby a side chain of a reactive lysine residue in the binding site of the
catalytic antibody
reacts with the reactive group Z to form an irreversible covalent bond.
2. The antibody targeting compound of claim 1, wherein said catalytic
aldolase
antibody is antibody 38C2.
3. The antibody targeting compound of claim 1 or 2, wherein said antibody
is full
length.
4. The antibody targeting compound of claim 1 or 2, wherein said antibody
is a
fragment of a full length antibody, and may be selected from the group
consisting of Fab, Fab'
F(ab')2, Fv and sFv.
5. The antibody targeting compound of any one of claims 1-4, wherein said
antibody is a human antibody, humanized antibody or chimeric human antibody.

68


6. The antibody targeting compound of any one of claims 1-5, wherein the X
group of said linker comprises a linear stretch of between 5-100 atoms.
7. The antibody targeting compound of claim 6, wherein the X group of said
linker comprises one or more groups selected from alkyl, alkenyl, alkynyl,
oxoalkyl,
oxoalkenyl, oxoalkynyl, aminoalkyl, aminoalkenyl, aminoalkynyl, sulfoalkyl,
sulfoalkenyl,
sulfoalkynyl group, phosphoalkyl, phosphoalkenyl, and phosphoalkynyl.
8. The antibody targeting compound of claim 6, wherein the X group of said
linker comprises a repeating ether unit of between 2-100 units.
9. The compound of any one of claims 6-8, wherein the X-group of said
linker is
branched and attached to different targeting agents.
10. The antibody targeting compound of any one of claims 1-9, wherein said
one
or more ring structures includes one or more six membered rings of the formula
Image
wherein each of A, B, C, D or W are independently selected from the group
consisting of C
and N.
11. The antibody targeting compound of claim 10, wherein the Y group
comprises
phenyl.
12. The antibody targeting compound of any one of claims 1-11, further
comprising a therapeutic drug covalently attached to the X group.
13. The antibody targeting compound of any one of claims 1-12, wherein said

targeting agent is a peptide.
14. A method of producing the antibody targeting compound of any one of
claims
1-13, said method comprising (1) linking the targeting agent to the connecting
chain of the

69


linker, and (2) irreversibly covalently linking said targeting agent to the
combining site of the
catalytic antibody via said linker.
15. The method of claim 14, wherein the catalytic aldolase antibody is
antibody
38C2.
16. Use of the antibody targeting compound of any one of claims 1-13 for
delivering a biological activity to cells, tissue extracellular matrix
biomolecule or a
biomolecule in the fluid of an individual, wherein said antibody targeting
compound is
specific for said cells, tissue extracellular matrix biomolecule or fluid
biomolecule and
wherein said antibody targeting compound comprises a biological activity.
17. The antibody targeting compound of any one of claims 1-13 for use in
delivering a biological activity to cells, tissue extracellular matrix
biomolecule or a
biomolecule in the fluid of an individual, wherein said antibody targeting
compound is
specific for said cells, tissue extracellular matrix biomolecule or fluid
biomolecule and
wherein said antibody targeting compound comprises a biological activity.
18. Use of a therapeutically effective amount of the antibody targeting
compound
of any one of claims 1-13 for treating or preventing a disease or condition in
an individual,
wherein:
said disease or condition involves cells, tissue or fluid that expresses a
target
molecule,
said antibody targeting compound is specific for said target molecule, and
said targeting compound comprises an agent effective against the disease or
condition.
19. The use of claim 18, wherein said agent is a cytokine, a toxin, a drug,
a nucleic
acid or an isotope.



20. The use of claim 18 or 19, wherein said disease or condition is an
infection and
said target molecule is expressed by a microbial agent or virus.
21. The use of any one of claims 18-20, wherein said compound is for in
vivo
administration.
22. The use of claim 21, wherein said compound is for topical
administration.
23. A therapeutically effective amount of the antibody targeting compound
of any
one of claims 1-13 for use in treating or preventing a disease or condition in
an individual,
wherein:
said disease or condition involves cells, tissue or fluid that expresses a
target
molecule,
said antibody targeting compound is specific for said target molecule, and
said targeting compound comprises an agent effective against the disease or
condition.
24. The compound of claim 23, wherein said agent is a cytokine, a toxin, a
drug, a
nucleic acid or an isotope.
25. The compound of claim 23 or 24, wherein said disease or condition is an

infection and said target molecule is expressed by a microbial agent or virus.
26. The compound of any one of claims 23-25, wherein said compound is for
in
vivo administration.
27. The compound of claim 26, wherein said compound is for topical
administration.
28. Use of the antibody targeting compound of any one of claims 1-13 for
imaging
cells or tissue in an individual wherein said cells or tissue expresses a
target molecule,
wherein said compound is linked to a detectable label.

71

29. The antibody targeting compound of any one of claims 1-13 for use in
imaging
cells or tissue in an individual wherein said cells or tissue expresses a
target molecule,
wherein said compound is linked to a detectable label.
30. Use of an effective amount of the antibody targeting compound of any
one of
claims 1-13 for reducing the infectivity of microbial cells or viral particles
present on a
surface, wherein said antibody targeting compound comprises a targeting agent
specific for a
receptor on said microbial cells or virus particles.

72

Description

Note: Descriptions are shown in the official language in which they were submitted.


CA 02464472 2004-04-20
WO 03/059251
PCT/US02/33991
ANTIBODY TARGETING COMPOUNDS
BACKGROUND OF THE INVENTION
[0001] The invention relates to compounds for targeting biological
molecules and
methods of making and using the compounds. Conventionally developed
pharmaceutical drugs and biological effector molecules are often of limited
use in
therapy because of high toxicity. Various approaches have been used over the
years to
improve the therapeutic index of such drugs orieffectors. One approach has
been to
couple a drug or effector to a ligand targeting agent such as an antibody. In
this case,
the antibody is used to change the distribution of drug or effector such that
more of it
can localize where it is most needed in vivo. Improved targeting of small
molecular
weight drugs or effectors has been achieved by complexing the drug or effector
with a
large molecular weight compound. For example, European Patent EP 217577
discloses
that increased half life and targeting by an agent is achieved by forming
complexes in
vivo between hapten-modified agents and anti-hapten antibodies. Similarly,
International Patent Application Publication WO 98/22141 discloses conjugates
of
therapeutic agents and haptens. The conjugates are administered to a subject
and
circulate in the blood stream of the subject. Circulating conjugates are
recognized and
bound by existing antibodies in the subject. Also, Shokat and Schultz (J. Am.
Chem.
Soc., 1991, 113:1862-1864) have disclosed a process for redirecting the immune

response using a process referred to as ligand-mediated immunogenicity.
According to
this teaching, an invariant antigen is complexed with a specific ligand and
administered
to a subject. The complexed invariant antigen then binds naturally occurring
antibodies
present in the subject.
BRIEF SUMMARY OF THE INVENTION
[0002] The present invention provides antibody targeting compounds with
unique
specificity and biological properties which are useful in many applications.
The
antibody targeting compounds of the invention comprise one or more targeting
agents
or biological agents or both covalently or noncovalently linked to an antibody
1

CA 02464472 2004-04-20
WO 03/059251
PCT/US02/33991
combining site. A linear or branched linker is preferably used in covalent and
non-
covalent linkage. Chemical characteristics of the linker are disclosed.
Depending on
the circumstances, the antibody specificity of the combining site may be
modified or
eliminated following covalent or noncovalent linking to the targeting or
biological
agent. In some embodiments, the antigen binding specificity of the antibody
before
covalent linkage may be substantially retained after covalent linkage.
[0003] The antibody targeting compound confers various benefits over the
components themselves. For example, the antibody portion of the compound may
generally extend the half-life of a smaller sized targeting or biological
agent in vivo.
Also, the biological potency or other biological feature of a particular
targeting or
biological agent may be modified by the addition of effector function(s)
provided by
the antibody portion of the compound (e.g., complement mediated effector
functions).
In addition, the targeting agent or binding agent, through its increased size
conferred by
linkage to the antibody, may enable the targeting agent to function in new
capacities.
[0004] In some embodiments, the targeting agent of the compound can bind to
a
non-immunoglobulin target molecule or to an immunoglobulin target molecule
outside
of the immunoglobulin combining site. Thus, in these embodiments, the
targeting
agent is specific for a non-antibody or is specific for an antibody but binds
to the
antibody outside its combining site. In a preferred approach, a catalytic
antibody can be
modified into a compound that binds specifically to a biomolecule. The
antibody
portion of the antibody targeting compounds can include whole antibody or
unique
antibody fragments and may have sequence derived from various animal species
such
as a non-human immunoglobulin or human immunoglobulin, the latter including a
human antibody, humanized antibody or human chimeric antibody.
[0005] Also provided are methods of producing antibody targeting compounds
of
the invention. In one embodiment, an agent-linker compound comprising a
targeting
agent and/or a biological agent is linked to a linker that comprises a
reactive group for
covalent reaction with the combining site of the antibody. In another
approach, an
antibody-linker compound is prepared where the linker includes a reactive
group for
2

CA 02464472 2004-04-20
WO 03/059251
PCT/US02/33991
reaction with said one or more targeting agents or biological agents. In yet
another
approach, the agents and the antibody can each be linked to a linkers with
compatible
reactive groups so that the antibody targeting compound forms when the two
linkers
covalently bond.
[0006] Further provided are agent-linker compounds comprising a targeting
agent,
biological agent or both that can be covalently linked to the combining site
of an
antibody. In some embodiments, the linker includes a reactive group for
covalently
linking the targeting agent to the combining site of the antibody. Linkage to
the
antibody combining site may be to a side chain of a reactive amino acid in the

combining site. In some embodiments, the reactive amino acid is a lysine while
the
linker reactive group is a ketone, a diketone, a beta lactam, a succinimide
active ester,
haloketone, a lactone, an anhydride, an epoxide, an aldehyde, a halide, a
sulfonate, a
phosphonate, a guanidine, an amidine, an imine, an eneamine, a ketal, a
acetal, or a
maleimide.
[0007] Various chemical features of the agent-linker compound are
described. In
one embodiment, the linker has the general formula X ¨ Y ¨ Z wherein X is a
linear or
branched connecting chain of atoms comprising any of C, H, N, 0, P, S, Si, F,
Cl, Br,
and I, or a salt thereof, and comprising a repeating ether unit of between 2-
100 units; Y
is optional and is a single or fused 5 or 6 membered homo- or heterocarbocylic

saturated or unsaturated ring located within 1-20 atoms of Z; and Z is a
reactive group
for covalently linking the one or more targeting agents to a side chain of a
reactive
amino acid in the combining site of the antibody. The targeting agent may be
linked to
X or Y or to X and Y when more than one targeting agent or biological agent is

included in the targeting agent-linker compound.
[0008] Yet further provided are targeting agent-linker-antigen compounds
for
noncovalently linking to the combining site of an antibody. These compounds
include
two or more targeting agents, two or more biological agents or at least two
agents, one
of which is a targeting agent and another a biological agent. The agents are
covalently
3

CA 02464472 2004-04-20
WO 03/059251
PCT/US02/33991
linked via a linker to an antigen recognized by the antibody. Various chemical
features
of the linker and antigen are disclosed.
[00091 Still further provided are methods of modifying an antibody which
exhibits
low or nondetectable binding affinity for a particular target molecule so that
the
antibody has increased binding specificity for the particular target molecule.
In one
embodiment, one or more targeting agents or biological agents specific for the

particular target molecule are covalently linked to the combining site of the
antibody to
generate an antibody targeting compound. The agents are linked in such a way
as to
retain their ability to bind the particular target molecule. In some such
embodiments,
the antibody prior to covalent linking possesses an affinity for the target
molecule of
less than about 1 x iO moles/liter. After covalent linking, the targeting
compound
may exhibit an affinity for the target molecule of greater than about 1 x 10-6
moles/liter.
100101 Additionally provided are methods of altering at least one physical
or
biological characteristic of a targeting agent or biological agent. In one
embodiment,
the agent is covalently linked to the combining site of an antibody to
generate an
antibody targeting compound. Methods are also provided for modifying one or
more
physical or biological properties of the antibody targeting compounds by
modifying one
or more chemical characteristics of the linker. In some embodiments, the
physical or
biological properties modified include pharmacokinetics, pharrnacodynamics,
irnmunogenicity, binding affinity, susceptibility to degradation, solubility,
lipophilicity,
hydrophilicity, hydrophobicity, stability, and rigidity.
100111 Also provided are methods of delivering a biological activity to
cells, an
extracellular matrix biomolecule or a fluid biomolecule of an individual. In
one
approach an antigen targeting compound of the invention which is biologically
active
and is specific for the cells, extracellular matrix biomolecule or fluid
biomolecule is
administered to the individual. In another approach, an agent-linker-antigen
compound
of the invention, specific for cells, tissue extracellular matrix biomolecule
or fluid
biomolecule, and an antibody specific for the antigen are separately
administered to the
4

CA 02464472 2004-04-20
WO 03/059251
PCT/US02/33991
individual and the antibody targeting agent forms in vivo when the agent-
linker-antigen
compound non-covalently associates with the antibody combining site.
[0012] Further provided are methods treating or preventing a disease or
condition
in an individual wherein the disease or condition involves cells, tissue or
fluid that
expresses a target molecule. In one approach, a therapeutically effective
amount of an
antibody targeting compound of the invention is administered to the
individual. In
another approach, a therapeutically effective amount of an agent-linker-
antigen
compound of the invention, and an antibody specific for the antigen are
separately
administered to the individual and the antibody targeting agent forms in vivo
when the
agent-linker-antigen compound non-covalently associates with the antibody
combining
site. In both approaches, the antibody targeting compound or agent-linker-
antigen
compound is specific for the target molecule, and the compound or antibody
comprises
a biological activity effective against the disease or condition.
[0013] Still further provided are methods of imaging cells or extracellular
matrix in
an individual wherein the cells or extracellular matrix express a target
molecule. In one
approach, an antibody targeting compound of the invention is linked to a
detectable
label and administered to the individual. In another approach an agent-linker-
antigen
compound and an antibody specific for the antigen are separately administered
to the
individual and the antibody targeting agent forms in vivo when the agent-
linker-antigen
compound non-covalently associates with the antibody combining site. In both
approaches, the label may be linked to the antibody, the targeting agent
and/or
biological agent.
[0014] Additionally provided are methods of reducing the infectivity of
microbial
cells or viral particles present on a surface. According to these methods, the
surface is
contacted with an effective amount of an antibody targeting compound of the
invention,
wherein the antibody targeting compound comprises a targeting agent or
biological
agent specific for a receptor on said microbial cells or virus particles.

CA 02464472 2004-04-20
WO 03/059251
PCT/US02/33991
[0015] Also provided are methods of screening a chemical library for
agonists or
antagonists of a receptor. The method includes linking individual members of
the
chemical library to the combining site of an antibody and then testing the
antibody
linked library for binding to the receptor or for inhibition of binding
between the
receptor and a ligand for the receptor.
[0016] Further provided are various immunoassays that use antibody
targeting
compounds of the invention. In one embodiment for detecting or measuring
analyte in
a sample, the invention comprises use of an antibody targeting compound of the

invention wherein the antibody specificity for the analyte results from the
targeting
agent, which is covalently linked to the antibody combining site. In another
embodiment involving a direct or indirect binding assay for determining the
presence of
an analyte using an antibody specific for the analyte, the invention comprises

determining the presence of the analyte using an antibody specific for the
analyte
wherein the antibody specificity results from a non-antibody targeting agent
specific for
the analyte that is linked to a reactive amino acid in the combining site of
the antibody.
[0017] Still further provided are methods of inhibiting or reducing the
ability of a
targeting agent or biological agent to cross a cell membrane. In these methods
an
antibody targeting compound is formed by covalently linking the combining site
of an
antibody that does not itself cross the cell membrane to the targeting agent
or biological
agent, wherein linkage of said antibody to said targeting agent or biological
agent
reduces or inhibits the ability of the agent to cross the cell membrane.
[0018] Additionally provided are methods of mediating intracellular
delivery of a
intracellularly active drug. In these methods, an antibody targeting compound
is
prepared wherein said compound includes one or more targeting agents or one or
more
biological agents or both covalently linked via a linker to the combining site
of the
antibody. The targeting agents or biological agents are characterized in that
they bind
to a cell receptor and mediate internalization of the agent. The antibody
targeting
compound also includes a drug that is active intracellularly. Intracellular
drug delivery
occurs when a cell expressing the receptor contacts the antibody targeting
compound.
6

CA 02464472 2013-01-17
=
28395-106
The contacting results in internalization of the antibody targeting agent and
delivery of said
drug intracellularly. In some embodiments, the intracellularly active drug is
a prodrug that
becomes active when said drug contacts an intracellular compartment. The
antibody targeting
compound may include an intracellular trafficking signal to direct the
internalized antibody
targeting compound to a particular intracellular compartment.
[00191 The invention further provides pharmaceutical compositions or
medicaments
that include an antibody targeting compound of the invention and a
pharmaceutically
acceptable carrier.
10019A1 Specific aspects of the invention include:
- an antibody targeting compound comprising at least one targeting agent at
least 500 daltons in size covalently linked to the combining site of a
catalytic aldolase
antibody via a linker of formula X-Y-Z, wherein the X group is a linear or
branched
connecting chain of atoms comprising any of C, H, N, 0, P, S, Si, F, Cl, Br,
and I, or a salt
thereof, the Y group is an optional recognition group comprising one or more
ring structures,
and the Z group is a reactive group:
o
o
Y IN4'''''''''
;
wherein Y refers to the Y group of the linker or X group if Y is absent;
wherein the targeting
agent is linked to the X group of the linker; and whereby a side chain of a
reactive lysine
residue in the binding site of the catalytic antibody reacts with the reactive
group Z to form an
irreversible covalent bond;
- a method of producing the antibody targeting compound as described herein,
said method comprising (1) linking the targeting agent to the connecting chain
of the linker,
and (2) irreversibly covalently linking said targeting agent to the combining
site of the
catalytic antibody via said linker;
7

CA 02464472 2013-01-17
=
28395-106
- use of the antibody targeting compound of the invention for delivering a
biological activity to cells, tissue extracellular matrix biomolecule or a
biomolecule in the
fluid of an individual, wherein said antibody targeting compound is specific
for said cells,
tissue extracellular matrix biomolecule or fluid biomolecule and wherein said
antibody
targeting compound comprises a biological activity;
- the antibody targeting compound of the invention for use in delivering a
biological activity to cells, tissue extracellular matrix biomolecule or a
biomolecule in the
fluid of an individual, wherein said antibody targeting compound is specific
for said cells,
tissue extracellular matrix biomolecule or fluid biomolecule and wherein said
antibody
targeting compound comprises a biological activity;
- use of a therapeutically effective amount of the antibody targeting compound

of the invention for treating or preventing a disease or condition in an
individual, wherein:
said disease or condition involves cells, tissue or fluid that expresses a
target molecule, said
antibody targeting compound is specific for said target molecule, and said
targeting compound
comprises an agent effective against the disease or condition;
- a therapeutically effective amount of the antibody targeting compound of the

invention for use in treating or preventing a disease or condition in an
individual, wherein:
said disease or condition involves cells, tissue or fluid that expresses a
target molecule, said
antibody targeting compound is specific for said target molecule, and said
targeting compound
comprises an agent effective against the disease or condition;
- use of the antibody targeting compound of the invention for imaging cells or

tissue in an individual wherein said cells or tissue expresses a target
molecule, wherein said
compound is linked to a detectable label;
- the antibody targeting compound of the invention for use in imaging cells or
tissue in an individual wherein said cells or tissue expresses a target
molecule, wherein said
compound is linked to a detectable label; and
7a

CA 02464472 2013-07-16
28395-106
- use of an effective amount of the antibody targeting compound of any one of
claims 1-13 for reducing the infectivity of microbial cells or viral particles
present on a
surface, wherein said antibody targeting compound comprises a targeting agent
specific for a
receptor on said microbial cells or virus particles.
BRIEF DESCRIPTION OF THE DRAWINGS
[0020] FIG. 1 shows exemplary integrin targeting agents of which
Panels A-E are
RGD peptidomimetic while Panel F is an RGD peptide. The core structures are
from the
following: U.S. Patent No. 6,335,330 (Panel A), U.S. Patent No. 5,693,636
(Panel B), U.S.
Patent No. 6,040,311 (Panel C), and U.S. Patent No. 6,001,117 (Panel E).
[0021] FIG. 2 shows a general scheme of a targeting agent-linker
compound with a
non-branched linker (Panel A) with specific embodiments in Panel B (SCS-873),
Panel C
(PST inhibitor diketo linker; compound 26), Panel D (TAK-799 diketo linker;
compound 27)
and Panel E (folate ligand dikone linker; compound 28).
[0022] FIG. 3 shows a general scheme of an embodiment of a targeting agent-
linker
compound with a branched linker and two identical targeting agents (Panel A)
with specific
embodiments in Panel B (integrin targeting agent diketo linker; compound 29),
and Panel C
(integrin targeting agent diketo linker; compound 30). The branch point is in
the connecting
chain portion of the linker.
[0023] FIG. 4 shows a general scheme of an embodiment of a targeting agent-
linker
compound with a branched linker and two different targeting agents (Panel A)
with a specific
embodiment in Panel B (integrin targeting and folate targeting agent
7b

CA 02464472 2004-04-20
WO 03/059251
PCT/US02/33991
diketo linker; compound 31). The branch point is in the connecting chain
portion of the
linker.
[0024] FIG. 5 shows a general scheme of an embodiment of a targeting agent-
linker compound with a branched linker and two different targeting agents
(Panel A)
with a specific embodiment in Panel B (integrin targeting agent diketo linker;

compound 32). The branch point is in the recognition group portion of the
linker.
[0025] FIG. 6 shows the structure of linker reactive groups. Structures A-C
form
reversible covalent bonds with reactive nucleophilic group (e.g. lysine or
cysteine side
chain) in the combining site of an antibody (structure A could form an
irreversible
covalent bond X is N and if RI and R3 form part of a cyclic structure). R1 and
R2 and
R3 in structures A-C represent substituents which can be C, H, N, 0, P, S, Si,
halogen
(F, Cl, Br, I) or a salt thereof. X is N, C, Si, or any other heteroatom.
These
substituents may also include a group such as an alkyl, alkenyl, alkynyl,
oxoalkyl,
oxoalkenyl, oxoalkynyl, aminoalkyl, aminoalkenyl, aminoalkynyl, sulfoalkyl,
sulfoalkenyl, or sulfoalkynyl group, phosphoalkyl, phosphoalkenyl,
phosphoalkynyl
group. R2 and R3 could be cyclic as exemplified in structures B and C while X
could be
a heteroatom. Structures D-G form nonreversible covalent bonds with reactive
nucleophilic group (e.g. lysine or cysteine side chain) in the combining site
of an
antibody. In these structures, R1 and R2 represent C, 0 N, halide and leaving
groups
such as mesyl or tosyl.
[0026] FIG. 7 shows various electrophiles suitable for reactive
modification with a
reactive amino acid side chain of an antibody. Key: (A) acyl beta-lactam; (B)
simple
diketone; (C) succinimide active ester; (D) maleimide; (E) haloacetamide with
linker;
(F) haloketone; (G) cyclohexyl diketone; and (H) aldehyde. R refers to other
structure
that may include a targeting agent, linker or antibody, while X refers to
halogen.
[0027] FIG. 8 shows the structure of linker recognition group (Y), situated
between
the reactive group portion and the connecting chain portion of the linker.
Panel A
8

CA 02464472 2004-04-20
WO 03/059251
PCT/US02/33991
shows the relationship of the recognition group Y within the linker (see FIG.
2). Panels
B-D show distance of Y from Z, substituents on the ring and ring member atoms.
[0028] FIG. 9 shows the structure of the linker connecting chain (X), which
directly attaches at one end to the targeting agent as shown in Panel A (see
FIG. 2).
Substituents R2 to R4 are C, H, N, 0, P, S, Si, halogen (F, Cl, Br, I) or a
salt thereof, and
may include a group such as an alkyl, alkenyl, alkynyl, oxoalkyl, oxoalkenyl,
oxoalkynyl, aminoalkyl, aminoalkenyl, aminoalkynyl, sulfoalkyl, sulfoalkenyl,
sulfoalkynyl group, phosphoalkyl, phosphoalkenyl, phosphoalkynyl as well as a
carbocyclic or heterocyclic mono or fused saturated or unsaturated ring
structure. Panel
B: R1 is 0 and R2 is C, H, N, 0, P, S, Si, halogen (F, Cl, Br, I) or a salt
thereof. In the
connecting chain in structures B and C, n, r or m is 1-100. In structures D
and E, n is
1, 2, 4, or more preferably is 3.
[0029] FIG. 10 shows Scheme 1, a synthetic scheme for the amine precursor
of
SCS-873, targeting agent 3 or SCS-amine. Key: (a) liBr3, CH2C12, -20 C, 2h;
(b)
DMF, rt to 80 C, 3h; (c) BnC0C1, sat. aq. NaHCO3, ether; (d) TBDPSiC1,
imidazole, DMF, 16h; (e) Pd(OAc)2, (o-to1)3P, i-Pr2EtN, CH3CH2CN, reflux, 3h;
(0
20% (w/w) Pd-C (10%), H2, Et0H-AcOH (1:1), 36h; (g) TBAF, THF, rt, lh; (h)
DEAD, PPh3, THF-benzene (3:1), 16h; (i) 20 % (w/w) Pd-C (10%), cyclohexene-i-
PrOH (1:1), 90 C, 12h; (j) i. aq. 2N NaOH, Me0H-THF (1:1), 16h, ii. TFAA,
anisole, CH2C12, 0 C, 2h.
[0030] FIG. 11 shows Scheme 2, a synthetic scheme for making Compound 4,
(R = Butoxycarboxyaminohexanoyl-derivative). Key: (a) DMF, rt; (b) EDC, HOBT,
DMF; (c) 0.01 M in DMSO, 130 C; (d) TFAA, anisole, dichloromethane; (e) DMF;
(f)
EDC, HOBT, DMF; (g) (i) step d, (ii) 2M NaOH, Me0H-THF (1:1).
[0031] FIG. 12 shows Scheme 3, a synthetic scheme for making compounds SCS-
873 and SCS-1655.
9

CA 02464472 2004-04-20
WO 03/059251
PCT/US02/33991
[0032] FIG. 13 shows Scheme 4, a synthetic scheme for making Compounds SCS-
864 and SCS-789. Key: (a) Et3N, DMF, rt, 16h.
[0033] FIG. 14 shows a scheme for forming a targeting agent-linker compound
using a linker with a maleimide-diketone reactive group.
DETAILED DESCRIPTION OF THE INVENTION
[0034] The present invention provides various antibody targeting compounds
in
which targeting agents and/or biological agents are covalently or
noncovalently linked
to the combining site of an antibody. When one or more targeting agents are
linked, at
least one of the targeting agents will be linked so that it can bind its
target. This may be
achieved by linking the targeting agent in a manner that does effect its
binding
specificity for the target and by sufficiently distancing the targeting agent
from the
antibody combining site so that it can bind its target without steric
hindrance by the
antibody. This may be achieved by using a suitable linker and linking strategy

discussed in more detail ahead.
[0035] When a biological agent is not also a targeting agent it is
preferred that the
antibody retain at least some antigen binding specificity following linkage to
one or
more biological agents. The antibody compound in which one or more biological
agents are linked to the antibody combining site may exhibit biological
activity due to a
linked biological agent if such agent is biologically active while linked to
the antibody.
This may be achieved by various strategies such as by linking the antibody
combining
site to a location on the biological agent that does not affect biological
activity.
Another strategy is to position the biological agent away from the antibody so
that the
biological agent can bind to another molecule necessary for activity without
steric
hindrance by the antibody. Other strategies for obtaining a biological
activity of one or
more biological agents linked to the antibody combining site are well known to
the
skilled artisan. In some embodiments, the biological activity of a biological
agent may
not be realized until the agent is released from the antibody combining site.
This may

CA 02464472 2004-04-20
WO 03/059251
PCT/US02/33991
be achieved in some embodiments though the aid of labile linkage as discussed
further
ahead.
[0036] In some embodiments, the native antigen binding specificity of the
antibody
which exists before covalent linkage will not be substantially modified
following
covalent linkage. In other words, the antibody compound resulting from
covalent
linkage of one or more targeting agents or one or more biological agents may
bind the
same antigens with a similar affinity as it did prior to covalent linkage. In
other
embodiments, the binding specificity of the antibody before covalent linkage
will be
substantially modified following covalent linkage. Substantially modified
antibody
binding specificity resulting from covalent linkage may be due to a
substantially
reduced ability of the covalently linked antibody to bind to an antigen or a
substantially
increased ability of the covalently linked antibody to bind to an antigen. In
some
embodiments, binding of the antigen binding site to antigen is sufficiently
reduced such
that the original antigen binding specificity of the antibody is effectively
eliminated. In
some embodiments, the antigen binding site to antigen is sufficiently reduced
such that
the original antigen binding specificity of the antibody is effectively
eliminated and
replaced with that of a targeting agent(s) covalently linked to the antibody
combining
site. In embodiments where the binding specificity of the antibody is
effectively
replaced with that of the targeting agent(s), the antibody, after covalent
linkage to the
targeting agent(s), exhibits an affinity for the target molecule of greater
than about 1 x
10-6 moles/liter.
[0037] Although not wishing to be bound by any theory, substantially
reduced
antibody binding to antigen may result from the targeting agent(s) or
biological agent(s)
sterically hindering the antigen from contacting the antibody combining site.
Alternatively, or in addition, substantially reduced antigen binding may
result if the
amino acid side chain of the antibody combining site modified by covalent
linkage was
important for binding to the antigen. Substantially increased antibody binding
to an
antigen may result when the targeting agent(s) or biological agent(s) do not
sterically
hinder the antigen from contacting the antibody combining site and amino acid
side
11

CA 02464472 2004-04-20
WO 03/059251
PCT/US02/33991
chain of the antibody combining site modified by covalent linkage was
important for
binding to the antigen.
[0038] The targeting compounds of the invention may comprise an antibody or
an
antibody fragment that has a single combining site such as Fab or Fab'
antibody
fragments. In such cases, the targeting agent will be linked to the single
combining site
of that antibody molecule. If an antibody or antibody fragment of a targeting
molecule
comprises two or more combining sites, at least one of the combining sites
will include
a covalently linked targeting agent. In some cases, all or most of the
combining sites of
an antibody can be covalently linked to a targeting agent. If multiple
combining sites of
an antibody are to be linked to targeting agents, the combining sites may all
have the
same targeting agent linked thereto or may have different targeting agents
linked to the
same antibody. It would be readily understood that one could covalently link
multiple
targeting agents to a single antibody combining site. Such multimeric
targeting agents
may be heteromultimeric or homomultimeric with respect to the specificity of
the
targeting agents in the multimer.
[0039] "Targeting agent" or "targeting component" as used herein refers to
a
moiety that recognizes, binds or adheres to a target moiety of a target
molecule located
for example in a cell, tissue (e.g. extracellular matrix), fluid, organism, or
subset
thereof. A targeting agent and its target molecule represent a binding pair of
molecules,
which interact with each other through any of a variety of molecular forces
including,
for example, ionic, covalent, hydrophobic, van der Waals, and hydrogen
bonding, so
that the pair have the property of binding specifically to each other.
Specific binding
means that the binding pair exhibit binding with each other under conditions
where they
do not bind to another molecule. Examples of binding pairs are biotin-avidin,
hormone-receptor, receptor-ligand, enzyme-substrate, lgG-protein A, antigen-
antibody,
and the like. The targeting agent and its cognate target molecule exhibit a
significant
association for each other. This association may be evaluated by determining
an
equilibrium association constant (or binding constant) according to methods
well
12

CA 02464472 2004-04-20
WO 03/059251
PCT/US02/33991
known in the art. Affinity is calculated as IQ =lcoff /kon (lcoff is the
dissociation rate
constant, kon is the association rate constant and IQ is the equilibrium
constant.
[0040] Affinity can be determined at equilibrium by measuring the fraction
bound
(r) of labeled ligand at various concentrations (c). The data are graphed
using the
Scatchard equation: r/c = K(n-r):
where
r = moles of bound ligand/mole of receptor at equilibrium;
c = free ligand concentration at equilibrium;
K = equilibrium association constant; and
n = number of ligand binding sites per receptor molecule
By graphical analysis, r/c is plotted on the Y-axis versus r on the X-axis
thus producing
a Scatchard plot. The affinity is the negative slope of the line. koff can be
determined
by competing bound labeled ligand with unlabeled excess ligand (see, e.g.,
U.S. Pat No.
6,316,409). The affinity of a targeting agent for its target molecule is
preferably at least
about 1 x 10-6 moles/liter, is more preferably at least about 1 x i0
moles/liter, is even
more preferably at least about 1 x 10-8 moles/liter, is yet even more
preferably at least
about 1 x 10-9 moles/liter, and is most preferably at least about 1 x 10-10
moles/liter.
[0041] Targeting agents include, but are not limited to, small molecule
organic
compounds of 5,000 daltons or less such as drugs, proteins, peptides,
peptidomimetics,
glycoproteins, proteoglycans, lipids glycolipids, phospholipids,
lipopolysaccharide,
nucleic acids, proteoglycans, carbohydrates, and the like. Targeting agents
may include
well known therapeutic compounds including anti-neoplastic agents. Anti-
neoplastic
targeting agents may include targpaclitaxel, daunorubicin, doxorubicin,
carminomycin,
4'-epiadriarnycin, 4-demethoxy-daunomycin, 11 -deoxydaunorubicin, 13-
deoxydaunorubicin, adriamycin-14-benzoate, adriamycin-14-octanoate, adriamycin-
14-
naphthaleneacetate, vinblastine, vincristine, mitomycin C, N-methyl mitomycin
C,
bleomycin A2, dideazatetrahydrofolic acid, aminopterin, methotrexate,
cholchicine and
cisplatin, and the like. Anti-microbial agents include aminoglycosides
including
gentamicin, antiviral compounds such as rifampicin, 3'-azido-3'-deoxythymidine
(AZT)
13

CA 02464472 2004-04-20
WO 03/059251
PCT/US02/33991
and acylovir, antifungal agents such as azoles including fluconazole, plyre
macrolides
such as amphotericin B, and candicidin, anti-parasitic compounds such as
antimonials,
and the like. Hormone targeting agents include toxins such as diphtheria
toxin,
cytokines such as CSF, GSF, GMCSF, TNF, erythropoietin, immunomodulators or
cytokines such as the interferons or interleukins, a neuropeptide,
reproductive hormone
such as HGH, FSH, or LH, thyroid hormone, neurotransmitters such as
acetylcholine,
and hormone receptors such as the estrogen receptor.
[0042] In some preferred embodiments, the targeting agent is not an
antibody. In
other preferred embodiments, the targeting agent is not a metal chelate.
Preferably, the
targeting agent is a small molecule as compared with a native immunoglobulin.
The
targeting agent, including any linking moiety necessary for covalently linking
the
targeting agent to an amino acid residue of the antibody combining site,
preferably is at
least about 300 daltons in size, and preferably may be at least about 400,
500, 600, 700,
800, 900, 1,000, 1,100, 1,200, 1,300, 1,400, 1,500, 1,600, 1,700, 1,800,
1,900, 2,000,
2,500, 3,000, 3,500, 4,000, 4,500 or even 5,000 daltons in size, with even
larger sizes
possible.
[0043] Suitable targeting agents in targeting compounds of the invention
can be a
protein or peptide. "Polypeptide", "peptide," and "protein" are used
interchangeably to
refer to a polymer of amino acid residues. As used herein, these terms apply
to amino
acid polymers in which one or more amino acid residue is an artificial
chemical
analogue of a corresponding naturally occurring amino acid. These terms also
apply to
naturally occurring amino acid polymers. Amino acids can be in the L or D form
as
long as the binding function of the peptide is maintained. Peptides can be of
variable
length, but are generally between about 4 and 200 amino acids in length.
Peptides may
be cyclic, having an intramolecular bond between two non-adjacent amino acids
within
the peptide, e.g., backbone to backbone, side-chain to backbone and side-chain
to side-
chain cyclization. Cyclic peptides can be prepared by methods well know in the
art.
See e.g., U.S. Pat. No. 6,013,625.
14

CA 02464472 2004-04-20
WO 03/059251
PCT/US02/33991
[0044] Protein or peptide targeting agents that exhibit binding activity
for a target
molecule are well known in the art. For example, a targeting agent may be a
viral
peptide cell fusion inhibitor. This may include the T-20 HIV-1 gp41 fusion
inhibitor
which targets fusion receptors on HIV infected cells (for T-20, see U.S.
Patent Nos.
6,281,331 and 6,015,881 to Kang et al.; Nagashima et al. J. Infectious
Diseases
183:1121, 2001; for other HIV inhibitors see U.S. Pat. No. 6020459 to Barney
and WO
0151673A2 to Jeffs et al), RSV cell fusion inhibitors (see WO 0164013A2 to
Antczak
and McKimm-Breschkin, Curr. Opin. Invest. Drugs 1:425-427, 2000 (VP-14637)),
pneumovirus genus cell fusion inhibitors (see WO 9938508A1 by Nitz et al.),
and the
like. Targeting agents also include peptide hormones or peptide hormone
analogues
such as LHRH, bombesin/gastrin releasing peptide, somatastatin (e.g., RC-121
octapeptide), and the like, which may be used to target any of a variety of
cancers
ovarian, mammary, prostate small cell of the lung, colorectal, gastric, and
pancreatic.
See, e.g., Schally et al., Eur. J. Endocrinology, 141:1-14, 1999.
[0045] Peptide targeting agents suitable for use in targeting compounds of
the
invention also may be identified using in vivo targeting of phage libraries
that display a
random library of peptide sequences (see, e.g., Arap et al., Nature Medicine,
2002
8(2):121-7; Arap et al., Proc. Natl. Acad. Sci. USA 2002 99(3):1527-1531;
Trepel et al.
Curr. Opin. Chem. Biol. 2002 6(3):399-404).
[0046] In some embodiments, the targeting agent is specific for an
integrin.
Integrins are heterodimeric transmembrane glycoprotein complexes that function
in
cellular adhesion events and signal transduction processes. Integrin a,133 is
expressed
on numerous cells and has been shown to mediate several biologically relevant
processes, including adhesion of osteoclasts to bone matrix, migration of
vascular
smooth muscle cells, and angiogenesis. Integrin av133 antagonists likely have
use in the
treatment of several human diseases, including diseases involving
neovascularization,
such as rheumatoid arthritis, cancer, and ocular diseases.

CA 02464472 2004-04-20
WO 03/059251
PCT/US02/33991
[0047] Suitable targeting agents for integrins include RGD peptides or
peptidomimetics or non-RGD peptides or peptidomimetics. As used herein,
reference
to "Arg-Gly-Asp peptide" or "RGD peptide" is intended to refer to a peptide
having one
or more Arg-Gly-Asp containing sequence which may function as a binding site
for a
receptor of the "Arg-Gly-Asp family of receptors", e.g., an integrin.
Integrins, which
comprise and alpha and a beta subunit, include numerous types including aiI3i,
a2I31,
a3I31, a413i, a5131, a6131, a7131, as131, a913i, ai1, a6(34, a4r37, an132,
aD132,0032,
am132,avP Lav133, avI35, av136, av138, axI32, a11d33, ainbi37, and the like.
The sequence RGD is
present in several matrix proteins and is the target for cell binding to
matrix by
integrins. Platelets contain a large amount of RGD-cell surface receptors of
the protein
GP IIb/IIIa, which is primarily responsible, through interaction with other
platelets and
with the endothelial surface of injured blood vessels, for the development of
coronary
artery thrombosis. The term RGD peptide also includes amino acids that are
functional
equivalents (e.g., RLD or KGD) thereof provided they interact with the same
RGD
receptor. Peptides containing RGD sequences can be synthesized from amino
acids by
means well known in the art, using, for example, an automated peptide
synthesizer,
such as those manufactured by Applied Biosystems, Inc., Foster City, Calif.
[0048] As used herein, "non-RGD" peptide refers to a peptide that is an
antagonist
or agonist of integrin binding to its ligand (e.g. fibronectin, vitronectin,
laminin,
collagen etc.) but does not involve an RGD binding site. Non-RGD integrin
peptides
are known for avi33 (see, e.g., U.S. Pat. Nos. 5,767,071 and 5,780,426) as
well as for
other integrins such as 041 (VLA-4), a4137 (see, e.g., U.S. Pat. Nos.
6,365,619; Chang
et al., Bioorganic & Medicinal Chem Lett, 12:159-163 (2002); Lin et al.,
Bioorganic &
Medicinal Chem Lett, 12:133-136 (2002)), and the like.
[0049] An integrin targeting agent may be a peptidomimetic agonist or
antagonist,
which preferably is a peptidomimetic agonist or antagonist of an RGD peptide
or non-
RGD peptide. As used herein, the term "peptidomimetic" is a compound
containing
non-peptidic structural elements that are capable of mimicking or antagonizing
the
biological action(s) of a natural parent peptide. A peptidomimetic of an RGD
peptide is
16

CA 02464472 2004-04-20
WO 03/059251
PCT/US02/33991
an organic molecule that retains similar peptide chain pharmacophore groups of
the
RGD amino acid sequence but lacks amino acids or peptide bonds in the binding
site
sequence. Likewise, a peptidomimetic of a non-RGD peptide is an organic
molecule
that retains similar peptide chain pharmacophore groups of the non-RGD binding
site
sequence but lacks amino acids or peptide bonds in the binding site sequence.
A
"pharmacophore" is a particular three-dimensional arrangement of functional
groups
that are required for a compound to produce a particular response or have a
desired
activity. The term "RGD peptidomimetic" is intended to refer to a compound
that
comprises a molecule containing the RGD pharmacophores supported by an
organic/non-peptide structure. It is understood that an RGD peptidomimetic (or
non-
RGD peptidomimetic) may be part of a larger molecule that itself includes
conventional
or modified amino acids linked by peptide bonds.
100501 RGD peptidomimetics are well known in the art, and have been
described
with respect to integrins such as GPIIb/IIIa, av133 and av135 (See, e.g.,
Miller et al., J.
Med. Chem. 2000, 43:22-26; and International Patent Publications WO 0110867,
WO
9915178, WO 9915170, WO 9815278, WO 9814192, WO 0035887, WO 9906049, WO
9724119 and WO 9600730; see also Kumar et al., Cancer Res. 61:2232-
2238(2000)).
Many such compounds are specific for more than one integrin. RGD
peptidomimetics
are generally based on a core or template (also referred to as "fibrinogen
receptor
antagonist template"), to which are linked by way of spacers to an acidic
group at one
end and a basic group at the other end of the core. The acidic group is
generally a
carboxylic acid functionality while the basic group is generally a N-
containing moiety
such as an amidine or guanidine. Typically, the core structure adds a form of
rigid
spacing between the acidic moiety and the basic nitrogen moiety, and contains
one or
more ring structures (e.g., pyridine, indazole, etc.) or amide bonds for this
purpose. For
a fibrinogen receptor antagonist, generally, about twelve to fifteen, more
preferably
thirteen or fourteen, intervening covalent bonds are present (via the shortest

intramolecular path) between the acidic group of the RGD peptidomimetic and a
nitrogen of the basic group. The number of intervening covalent bonds between
the
acidic and basic moiety is generally shorter, two to five, preferably three or
four, for a
17

CA 02464472 2004-04-20
WO 03/059251
PCT/US02/33991
vitronectin receptor antagonist. The particular core may be chosen to obtain
the proper
spacing between the acidic moiety of the fibrinogen antagonist template and
the
nitrogen atom of the pyridine. Generally, a fibrinogen antagonist will have an

intramolecular distance of about 16 angstroms (1.6 nm) between the acidic
moiety (e.g.,
the atom which gives up the proton or accepts the electron pair) and the basic
moiety
(e.g., which accepts a proton or donates an electron pair), while a
vitronectin antagonist
will have about 14 angstroms (1.4 nm) between the respective acidic and basic
centers.
Further description for converting from a fibrinogen receptor mimetic to a
vitronectin
receptor mimetic can be found in U.S. Pat. No. 6,159,964.
[0051] The peptidomimetic RGD core can comprise a 5-11 membered aromatic or
nonaromatic mono- or polycyclic ring system containing 0 to 6 double bonds,
and
containing 0 to 6 heteroatoms chosen from N, 0 and S. The ring system may be
unsubstituted or may be substituted on a carbon or nitrogen atom. Preferred
core
structures with suitable substituents useful for vitronectin binding include
monocyclic
and bicyclic groups, such as benzazapine described in WO 98/14192,
benzdiazapine
described in U.S. 6,239,168, and fused tricyclics described in U.S. 6,008,213.
100521 U.S. Pat. No. 6,159,964 contains an extensive list of references in
Table 1
of that document which disclose RGD peptidomimetic cores structures (referred
to as
fibrinogen templates) which can be used for prepraring RGD peptidomimetics.
Preferred vitronectin RGD and fibronectin RGD peptidomimetics are disclosed in
U.S.
Patent Nos. 6,335,330; 5,977,101; 6,088,213; 6,069,158; 6,191,304; 6,239,138;
6,159,964; 6,117,910; 6,117,866; 6,008,214; 6,127,359; 5,939,412; 5,693,636;
6,403,578; 6,387,895; 6,268,378; 6,218,387; 6,207,663; 6,011,045; 5,990,145;
6,399,620; 6,322,770; 6,017,925; 5,981,546; 5,952,341; 6,413,955; 6,340,679;
6313,119; 6,268,378; 6,211,184; 6,066,648; 5,843,906; 6,251,944; 5,952,381;
5,852,210; 5,811,441; 6,114,328; 5,849,736; 5,446,056; 5,756,441; 6,028,087;
6,037,343; 5,795,893; 5,726,192; 5,741,804; 5,470,849; 6,319,937; 6,172,256;
5,773,644; 6,028,223; 6,232, 308; 6,322,770; 5,760,028.
18

CA 02464472 2004-04-20
WO 03/059251
PCT/US02/33991
[0053] Exemplary RGD peptidomimetic Mtegrin targeting agents are shown
below
as compounds 1, 2, and 3 can be used for preparing an intregrin targeting
compound of
the present invention. In the three compounds, the linker is attached as
indicated to the
nitrogen of the seven membered ring. Other RGD peptidomimetic integrin
targeting
agents include compound 33, wherein P and L or carbon or nitrogen. The linker
may
be R1 or R2 while the R3 group includes a basic group such as an ¨NH group. In
some
embodiments, the R3 group is as shown in compounds 1, 2, or 33. In some
embodiments, the R3 group includes a heterocyclic group such a benzimidazole,
imidazole, pyridine group, or the like. In some such embodiments, the R3 group
is a
alkoxy group, such as a propoxy group or the like, that is substituted with a
heterocyclyl group that is substituted with an alkylamine group, such as a
methylamino
group or the like, whereas in other embodiments, the R3 group is an alkoxy
group, such
as a propoxy group or the like, substituted with a heterocyclylamino group,
such as with
a pyridinylamino group or the like such as a 2-pyridinylamino group. In other
embodiments R3 is a group of formula ¨C(=0)Rb where Rb is selected from
¨N(alkyl)-
alkyl-heterocyclyl groups such as ¨N(Me)-CH2-benzimidazole groups and the
like.
N
0
CO2H
0
NT
46 N 0 , NH
GO2H
2
N is N
0
02 H
3
19

CA 02464472 2004-04-20
WO 03/059251
PCT/US02/33991
O
n n = 0-5
R34
CO2R4
rs2
31
[0054] Other exemplary integrin peptidomimetic targeting agents and a
peptide
targeting agent are shown in FIG. I. The linker may be any of R1, R2, R3,
while R4 may
be a linker or a hydrolyzable group such as alkyl, alkenyl, alkynyl, oxoalkyl,

oxoalkenyl, oxoalkynyl, aminoalkyl, aminoalkenyl, aminoalkynyl, sulfoalkyl,
sulfoalkenyl, or sulfoalkynyl group, phosphoalkyl, phosphoalkenyl,
phosphoalkynyl
group, and the like. One of skill in the art will readily appreciate that
other integrin
agonist and antagonist mimetics can also be used in targeting compounds of the
present
invention.
[0055] The target molecule to which the targeting agent of the targeting
compound
binds is preferably a non-immunoglobulin molecule or is an immunoglobulin
molecule
where the target moiety is outside the immunoglobulin combining site. It is
not
intended to exclude from the inventive compounds those targeting agents that
function
as antigens and, therefore, bind to an immunoglobulin combining site. Such
targeting
agents are included herein provided the targeting agents also bind to a non-
immunoglobulin molecule and/or a target moiety located outside the combining
site of
an immunoglobulin molecule. In general, the target molecule can be any type of

molecule including organic, inorganic, protein, lipid, carbohydrate, nucleic
acid and the
like.
[0056] The target molecule is preferably a biomolecule such as a protein,
carbohydrate, lipid or nucleic acid. The target molecule can be associated
with a cell

CA 02464472 2004-04-20
WO 03/059251
PCT/US02/33991
("cell surface expressed"), or other particle ("particle surface expressed")
such as a
virus, or may be extracellular. If associated with a cell or particle, the
target molecule
is preferably expressed on the surface of the cell or particle in a manner
that allows the
targeting agent of the targeting compound to make contact with the surface
receptor
from the fluid phase of the body.
[0057] In some preferred embodiments, the target molecule is predominantly
or
exclusively associated with a pathological condition or diseased cell, tissue
or fluid.
Thus, the targeting agent of a present antibody targeting compound can be used
to
deliver the targeting compound to a diseased tissue by targeting the cell, an
extracellular matrix biomolecule or a fluid biomolecule. Exemplary target
molecules
disclosed hereinafter in the Examples include integrins (Example 1), cytokine
receptors
(Examples 2, 3 and 7), cytokines (Example 4), vitamin receptors (Example 5),
cell
surface enzymes (Example 6), and HIV-1 virus and HIV-1 virus infected cells
(Examples 8 and 11), and the like.
[0058] In other preferred embodiments, the target molecule is associated
with an
infectious agent and is expressed on the surface of a microbial cell or on the
surface of
a viral particle. As such, antibody targeting compositions in which the
targeting agent
can bind to the cell surface expressed or particle expressed infectious agent
can be used
as an anti-microbial, by targeting microbial agents inside the body or on the
surface
(e.g., skin) of an individual. In the latter case, the invention compound can
be applied
topically.
[0059] Antibody targeting agents specific for a microbial target molecule
also can
be used as an anti-microbial agent in vitro. Accordingly, a method of reducing
the
infectivity of microbial cells or viral particles present on a surface is
provided. Some
methods include contacting the surface of a microbial cell or viral particle
with an
effective amount of the invention targeting compound. The targeting compound
in
such methods includes .a targeting agent specific for a receptor on the
microbial cell or
virus particle. Applicable surfaces are any surfaces in vitro such as a
counter top,
condom, and the like.
21

CA 02464472 2004-04-20
WO 03/059251 PCT/US02/33991
[0060] Another preferred target molecule for targeting molecules of the
invention
is prostate specific antigen (PSA), a serine protease that has been implicated
in a variety
of disease states including prostate cancer, breast cancer and bone
metastasis. Specific
inhibitors of PSA which bind to the active site of PSA are known. See
Adlington et al.,
J. Med. Chem., 2001, 44:1491-1508 and WO 98/25895 to Anderson. A specific
inhibitor of PST is shown below as compound 34.
0
HO 411
0 10 0
0 0 OH
0
0
=
34
[0061] A targeting agent, in addition to its ability to bind a target
molecule, may be
characterized in having one or more biological activities, each activity
characterized as
a detectable biological affect on the functioning of a cell organ or organism.
Thus, in
addition to being a targeting agent, such compounds can be considered
biological
agents. For example, the integrin targeting agents shown as compounds 1, 2, 3
and 33
above not only target an integrin, but have integrin antagonist biological
activity. In
some embodiments, however, a targeting agent may be a pure binding agent
without
biological activity.
[0062] The targeting compounds of the invention include a targeting agent
that is
=
covalently linked to a combining site of an antibody. Such targeting compounds
may
have one or more biological activities associated with the targeting compound.
The
biological activity may be an inherent feature of the targeting agent itself
or may be
provided by a biological agent distinct from the targeting agent in the
targeting
compound. The biological agent may be associated covalently or non-covalently
with
22

CA 02464472 2004-04-20
WO 03/059251
PCT/US02/33991
the other molecules or portions of the targeting compound, although covalent
linkage is
preferred. The biological agent may be linked to either the targeting agent,
the
antibody, or both by means well known in the art. For example, see Kiaris et
al., Eur. J.
Cancer 37:620-628 (2001) and Schally etal. Eur. J. Endocrin. 141:1-14 (1989),
which
describe various conjugates between peptide hormone targeting agents and
doxorubicin.
See also, Canevari et al., Ann Oncol 1994 Oct;5(8):698-701; Rihova, Folia
Microbiol
(Praha) 1995;40(4):367-84; Vitetta, Princess Takamatsu Symp 1988;19:333-40;
and
Ghose et al., Crit Rev Ther Drug Carrier Syst 1987;3(4):263-359. Thus, in some

embodiments, the antibody-targeting agent targeting compounds of the invention
may
include a functional component in the form of a targeting agent that has
inherent
biological activity. In such embodiments, the targeting agent is linked to a
combining
site of the antibody or antibody fragment and the targeting agent is the
functional
component that exhibits the biological activity. In other embodiments, the
targeting
compound includes a targeting agent linked to a combining site of an antibody
or
antibody fragment, and also includes a separate functional component that is
preferably
attached or linked to the targeting compound through a covalent bond.
[0063] A targeting agent or biological agent can be linked to an antibody
targeting
compound of the invention using a linkage that is labile under certain
conditions. The
labile linkage may be between the antibody and the targeting agent or
biological agent,
while if a linker is present, the labile linkage may be between the antibody
and the
linker, the targeting agent or biological agent and the linker, within the
linker, or
combinations thereof.
[0064] Labile linkers include, reversible covalent bonds, pH sensitive
linkages
(acid or base sensitive), enzyme sensitive linkages, degradation sensitive
linkers,
photosensitive linkers, sand the like, and combinations thereof. These
features are also
characteristic of a prodrug which can be considered as a type of labile
linker. A variety
of labile linkers have been previously designed. For example, prodrugs can be
formed
using compounds having carboxylic acid moieties that slowly degrade by
hydrolysis as
described in U.S. Patent No. 5,498,729.
23

CA 02464472 2004-04-20
WO 03/059251
PCT/US02/33991
[0065] The particular design of a labile linker may be used to direct
release of the
biological agent after it has reached the intended target. For example, a
linkage may be
designed to direct release in a particular intracellular compartment or in an
extracellular
compartment in which antibody targeting compounds may accumulate. An acid-
labile
linker such as a cis-aconitic acid linker can take advantage of the acidic
environment of
different intracellular compartments such as the endosomes encountered during
receptor mediated endocytosis and the lysosomes. See Shen et al., Biochem.
Biophys.
Res. Commun. (1981) 102:1048-1054; Yang et al., J. Natl. Canc. Inst. (1988)
80: 1154-
1159. A peptide spacer arm located within or at the ends of a linker can be
used to
effect release of a targeting agent or biological agent by the action of a
peptidase such
as a lysosomal peptidase. See e.g., Trouet et al., Proc. Natl. Acad. Sci.
(1982) 79: 626-
629.
[0066] Particular targeting agents may or may not possess biological
activity
depending on the context of their use. For example, the therapeutic drug
doxorubicin,
which is a DNA intercalator, can be a targeting agent for double stranded DNA
when
the drug is covalently linked to an antibody and applied to DNA in a cell-free
form.
Doxorubicin, however, may not be considered a targeting agent with respect to
a cell
while the drug is covalently linked to an antibody unless the compound can be
taken up
by the cell. In the latter case, doxorubicin may have biological activity
following
uptake if the drug can access DNA in the cell nucleus.
[0067] Biological agent functional components include, but are not limited
to,
small molecule drugs (a pharmaceutical organic compound of about 5,000 daltons
or
less), organic molecules, proteins, peptides, peptidomimetics, glycoproteins,
proteoglycans, lipids glycolipids, phospholipids, lipopolysaccharides, nucleic
acids,
proteoglycans, carbohydrates, and the like. Biological agents may be anti-
neoplastic,
anti-microbial, a hormone, an effector, and the like. Such compounds include
well
known therapeutic compounds such as the anti-neoplastic agents paclitaxel,
daunorubicin, doxorubicin, carrninomycin, 4'-epiadriamycin, 4-demethoxy-
daunomycin, 11 -deoxydaunorubicin, 13-deoxydaunorubicin, adriamycin-14-
benzoate,
24

CA 02464472 2004-04-20
WO 03/059251
PCT/US02/33991
adriamycin-14-octanoate, adriamycin-14-naphthaleneacetate, vinblastine,
vincristine,
mitomycin C, N-methyl mitomycin C, bleomycin A2, dideazatetrahydrofolic acid,
aminopterin, methotrexate, cholchicine and cisplatin, and the like. Anti-
microbial
agents include aminoglycosides including gentamicin, antiviral compounds such
as
rifampicin, 3'-azido-3'-deoxythymidine (AZT) and acylovir, antifungal agents
such as
azoles including fluconazole, plyre macrolides such as amphotericin B, and
candicidin,
anti-parasitic compounds such as antimonials, and the like. Hormones may
include
toxins such as diphtheria toxin, cytokines such as CSF, GSF, GMCSF, TNF,
erythropoietin, immunomodulators or cytokines such as the interferons or
interleukins,
a neuropeptide, reproductive hormone such as HGH, FSH, or LH, thyroid hormone,

neurotransmitters such as acetylcholine, hormone receptors such as the
estrogen
receptor. Also included are non-steroidal anti-inflammatories such as
indomethacin,
salicylic acid acetate, ibuprofen, sulindac, piroxicam, and naproxen, and
anesthetics or
analgesics. Also included are radioisotopes such as those useful for imaging
as well as
for therapy.
[0068] Biological agent functional components for use in the targeting
compounds
of the invention can be naturally occurring or synthetic. Biological agents
can be
biologically active in their native state, or be biologically inactive or in a
latent
precursor state and acquire biological or therapeutic activity when a portion
of the
biological agent is hydrolyzed, cleaved or is otherwise modified. The prodrug
can be
delivered at the surface of a cell or intracellulary using antibody targeting
compounds
of the invention where it can then be activated. In this regard, the
biological agent can
be a "prodrug," meaning that prodrug molecules capable of being converted to
drugs
(active therapeutic compounds) by certain chemical or enzymatic modifications
of their
structure. In the prodrug approach, site-specific drug delivery can be
obtained from
tissue-specific activation of a prodrug, which is the result of metabolism by
an enzyme
that is either unique for the tissue or present at a higher concentration
(compared with
other tissues); thus, it activates the prodrug more efficiently.

CA 02464472 2004-04-20
WO 03/059251
PCT/US02/33991
[0069] Photodynamic treatment may be used to activate a prodrug by cleaving
a
photosenitive linker or by activating a photoresponsive enzyme (acyl enzyme
hydrolysis) as described previously (see U.S. Patent No. 5,114,851 and
5,218,137).
Photodynamic treatment also may be used to rapidly inactivate a drug in sites
where the
drug activity is not desired (e.g. in non-target tissues). Various means of
covalently
modifying a drug to form a prodrug are well known in the art.
[0070] Targeting agents may be covalently linked to the antibody combining
site
directly or through the aid of a linker. An appropriate linker can be chosen
to provide
sufficient distance between the targeting agent and the antibody combining
site in order
for the targeting agent to be able to bind to its target molecule. This
distance depends
on several factors including, for example, the distance from the outermost
surface of the
antibody combining site to the reactive side chain in the combining site, and
the nature
of the targeting agent. Generally, the linker will be between about 5 to 10
angstroms
(0.5 to 1 nm) in length, with 10 or more angstroms (1.0 nm) being more
preferred,
although shorter linkers of about 3 angstroms (0.3 nm) in length may be
sufficient if the
amino acid side chain is very near to the outermost portion of the combining
site and/or
the targeting agent or biological agent includes a segment that can function
as a part of
a linker.
[0071] Linker length may also be viewed in terms of the number of linear
atoms
(cyclic moieties such as aromatic rings and the like to be counted by taking
the shortest
route). Linker length under this measure is generally about 10 to 200 atoms
and more
typically about 30 or more atoms, although shorter linkers of two or more
atoms may be
sufficient if the reactive amino acid side chain is very near to the outermost
portion of
the combining site. Generally, linkers with a linear stretch of at least about
9 atoms are
sufficient. Other linker considerations include effect on physical or
pharmacokinetic
properties of the resulting targeting compound or targeting agent-linker,
solubility,
lipophilicity, hydrophilicity, hydrophobicity, stability (more or less stable
as well as
planned degradation), rigidity, flexibility, immunogenicity, modulation of
antibody
26

CA 02464472 2004-04-20
WO 03/059251
PCT/US02/33991
binding, chemical compatibility with targeting agent, ability to be
incorporated into a
micelle or liposome, and the like.
[0072] In targeting compounds where a linker is present between the
antibody
combining site, the targeting agent may be prepared by several approaches. In
one
approach, a targeting agent-linker compound and/or biological agent-linker
compound
is synthesized with a linker that includes one or more reactive groups
designed for
covalent reaction with a side chain of an amino acid in the combining site of
an
antibody. The agent-linker compound and antibody are combined under conditions

where the linker reactive group forms a covalent bond with the amino acid side
chain.
[0073] In another approach, linking can be achieved by synthesizing an
antibody-
linker compound comprising an antibody and a linker wherein the linker
includes one
or more reactive groups designed for covalent reaction with an appropriate
chemical
moiety of the targeting agent or biological agent. The targeting agent or
biological
agent may need to be modified to provide the appropriate moiety for reaction
with the
linker reactive group. The antibody-linker and targeting agent and/or
biological agent
are combined under conditions where the linker reactive group covalently links
to the
targeting and/or biological agent.
[0074] A further approach for forming an antibody targeting compound of the
invention uses a dual linker design. In one embodiment, the an agent-linker
compound
is synthesized which comprises a targeting agent and/or a biological agent and
a linker
with a reactive group. An antibody-linker compound is synthesized which
comprises
an antibody and a linker with a chemical group susceptible to reactivity with
the
reactive group of the agent-linker of the first step. These two linker
containing
compounds are then combined under conditions whereby the linkers covalently
link,
forming the antibody targeting compound.
[0075] In another embodiment, an antibody-linker compound is synthesized
which
comprises an antibody and a linker with a reactive group. A targeting agent
and/or
biological agent-linker compound is prepared which comprises the agent and a
linker
27

CA 02464472 2004-04-20
WO 03/059251
PCT/US02/33991
with a chemical group susceptible to reactivity with the reactive group of the
antibody-
linker of the first step. These two linker containing compounds are then
combined
under conditions whereby the linkers covalently link, forming the antibody
targeting
compound. "Susceptible" as used herein with reference to a chemical moiety
indicates
that the chemical moiety will covalently bond with a compatible reactive
group. Thus,
an electrophilic group is susceptible to covalent bonding with a nucleophillic
group and
vice versa.
[0076] As discussed, the linker may be first conjugated to the targeting
agent and
then the targeting agent-linker conjugated to the antibody combining site.
Alternatively, the linker may be conjugated first to the antibody combining
site and the
antibody-linker conjugated to the targeting agent. Numerous means well known
in the
art can be used to attach a linker to the targeting agent or antibody
combining site.
Exemplary functional groups that can be involved in the linkage include, for
example,
esters, amides, ethers, phosphates, amino, keto, amidine, guanidine, imines,
eneamines,
phosphates, phosphonates, epoxides, aziridines, thioepoxides, masked or
protected
diketones (ketals for example), lactams, haloketones, aldehydes,
thiocarbamate,
thioamide, thioester, sulfide, disulfide, phosphoramide, sulfonamide, urea,
thioruea,
carbamate, carbonate, hydroxamide, and the like.
[0077] The linker includes any atom from the group C, H, N, 0, P, S, Si,
halogen
(F, Cl, Br, I) or a salt thereof. The linker also may include a group such as
an alkyl,
alkenyl, alkynyl, oxoalkyl, oxoalkenyl, oxoalkynyl, aminoalkyl, aminoalkenyl,
aminoalkynyl, sulfoalkyl, sulfoalkenyl, or sulfoalkynyl group, phosphoalkyl,
phosphoalkenyl, phosphoalkynyl group. The linker also may include one or more
ring
structures. As used herein a "ring structure" includes a carbocyclic homo or
hetero
mono or fused saturated or unsaturated ring structure. Combinations of the
above
groups and rings may also be present in the linkers of the targeting compounds
of the
invention.
28

CA 02464472 2004-04-20
WO 03/059251
PCT/US02/33991
[0078] The general design of a embodiment of a unbranched linker for use in
preparing targeting compounds of the present invention is shown in FIG. 2A.
The
linker is of the formula
X ¨ Y ¨ Z
Wherein X is a connecting chain, Y is a recognition group and Z is a reactive
group.
Figure 2B-E shows various targeting agent-linker compounds with the linker X,
Y and
Z portions identified. The linker may be linear or branched. In some
embodiments, the
linker has a linear stretch of between 5-200 or 10-200 atoms although in other

embodiments, longer linker lengths may be used. One or more targeting agents
may be
linked to X. In some embodiments, where more than one targeting agent is
linked and a
branched linker is used, some of the targeting agents may be linked to
different
branches of the linker. However, it should be understood that linkers used in
the
compounds of the invention may have one or more recognition groups, one or
more
reactive groups and one or more connecting chains and combinations thereof.
Connecting chains may branch from another connecting chain or from a
recognition
group.
[0079] The connecting chain X of the linker includes any atom from the
group C,
H, N, 0, P, S, Si, halogen (F, Cl, Br, I) or a salt thereof. X also may
include a group
such as an alkyl, alkenyl, alkynyl, oxoalkyl, oxoalkenyl, oxoalkynyl,
aminoalkyl,
aminoalkenyl, aminoalkynyl, sulfoalkyl, sulfoalkenyl, or sulfoalkynyl group,
phosphoalkyl, phosphoalkenyl, phosphoalkynyl group. In some embodiments, X may

include one or more ring structures. In a preferred embodiment, X includes a
repeating
ether unit of between 2-100 units. Various embodiments of X are shown in FIG.
9.
[0080] The recognition group Y of the linker is optional and if present is
located
between the reactive group and the connecting chain. In preferred embodiments,
Y is
located from 1-20 atoms from Z. Although not wishing to be bound by any
theory, it is
believed that the recognition group acts to properly position the reactive
group into the
antibody combining site so that it may react with a reactive amino acid side
chain.
29

CA 02464472 2004-04-20
WO 03/059251
PCT/US02/33991
FIG. 8 shows a variety of exemplary recognition groups with one or more homo
or
hetero ring structures of five or six atoms. Larger ring structures also may
be used.
One or more targeting agents may be linked to Y. In some embodiments, a linker
may
be used to link the targeting agent to Y. In embodiments where two or more
targeting
agents are used, one or more can be attached to both X and Y. More than one
targeting
agent also can be attached to Y.
[0081] The linker reactive group Z includes any nucleophilic or
electrophilic
group. In a preferred embodiment Z is capable of forming a covalent bond with
a
reactive side chain of an antibody. In some embodiments, Z includes one or
more
C=0, groups arranged to form a diketone, an acyl beta-lactam, an active ester,

haloketone, a cyclohexyl diketone group, an aldehyde or maleimide. Other
groups may
include lactone, anhydride, and alpha-haloacetamide or an epoxide. Exemplary
linker
electrophilic reactive groups that can covalently bond to a reactive
nucleophilic group
(e.g. lysine or cysteine side chain) in the combining site of an antibody
include acyl
beta-lactam, simple diketone, succinimide active ester, maleimide,
haloacetamide with
linker, haloketone, cyclohexyl diketone, aldehyde, amidine, guanidine, imine,
eneamine, phosphate, phosphonate, epoxide, aziridine, thioepoxide, a masked or

protected diketone (a ketal for example), lactam, sulfonate, and the like
masked CO
groups such as imine, ketal, acetal and any other known electrophilic group. A

preferred linker reactive group includes one or more C=0, groups arranged to
form a
acyl beta-lactam, simple diketone, succinimide active ester, maleimide,
haloacetamide
with linker, haloketone, cyclohexyl diketone, or aldehyde.
[0082] Z may be a group that forms a reversible or nonreversible covalent
bond. In
some embodiments, reversible covalent bonds may be formed using diketone Z
groups
such as those shown in FIG. 6. R1 and R2 and R3 in structures A-C of FIG. 6
represent
substituents which can be C, H, N, 0, P, S, Si, halogen (F, Cl, Br, I) or a
salt thereof.
These substituents also may include a group such as an alkyl, alkenyl,
alkynyl,
oxoalkyl, oxoalkenyl, oxoalkynyl, aminoalkyl, aminoalkenyl, aminoalkynyl,
sulfoalkyl,
sulfoalkenyl, or sulfoalkynyl group, phosphoalkyl, phosphoalkenyl,
phosphoalkynyl

CA 02464472 2004-04-20
WO 03/059251
PCT/US02/33991
group. R2 and R3 also could from a ring structure as exemplified in structures
B and C.
X in FIG. 6 could be a heteroatom. Other Z groups that form reversible
covalent bonds
include the diketone amidine, imine, and other reactive groups shown in
structures B
and G of FIG. 7. FIG. 7 also includes the structures of other preferred linker
reactive
groups.
[0083] Z reactive groups that form a nonreversible covalent bond with the
combining site of an antibody include structures D-G in FIG. 6 and structures
A, C and
D of FIG. 7. Such structures are useful for nonreversibly attaching a
targeting agent-
linker to a reactive nucleophilic group (e.g. lysine or cysteine side chain)
in the
combining site of an antibody.
[0084] It should be understood that the above described reversible and
nonreversible covalent linking chemistry can also be applied to link a
targeting agent
or biological agent to an antibody in the absence of a linker or to link a
targeting agent
or biological agent to a linker (e.g. to the connecting chain of the linker).
For example,
a targeting agent can be linked to a linker to form a targeting agent-linker
by placing a
suitable reactive group Z type element such as an appropriate nucleophilic or
electrophilic group on either the linker or the targeting agent and a suitable
reactive
moiety such as an amino or sulfhydral group on the other of the two.
[0085] A preferred linker for use in targeting compounds of the invention
and for
preparing targeting agent-linker compounds includes a 1,3-diketone reactive
group as
Z. Another preferred linker is one where the connecting chain X includes a
repeating
ether unit of between 2-100 units. Linkers in which the recognition group Y is
present
are preferred with Y located preferably between 1-20 atoms from the reactive
group Z.
Such a linker attached to the core of an integrin targeting RGD peptidomimetic
moiety
such as those described above, can have the structure 28 as shown below where
n is
from 1-100 or more and preferably is 1, 2, or 4, and more preferably is 3. In
some
embodiments, the linker is a repeating polymer such as polyethylene glycol.
31

CA 02464472 2004-04-20
WO 03/059251
PCT/US02/33991
=
in
[0086] The linker reactive group or similar such reactive group that may be
inherent in the targeting agent, is chosen for use with a particular antibody.
For
example, a chemical moiety for modification by an aldolase antibody may be a
ketone,
diketone, beta lactam, active ester haloketone, lactone, anhydride, maleimide,
alpha-
haloacetamide, cyclohexyl diketone, epoxide, aldehyde, amidine, guanidine,
imine,
eneamine, phosphate, phosphonate, epoxide, aziridine, thioepoxide, masked or
protected diketone (ketal for example), lactam, haloketone, aldehyde, and the
like. A
1,3-diketone configuration such as the diketone shown in Compound SCS-873 (see

below) or SCS-864 (see below), is especially preferred as a substrate for
modification
by an aldolase antibody.
00
(70, N':%=04VNWN
0
CO2H
Targeting Agent Linker with 1,3 Diketone Group
SCS873
32

CA 02464472 2004-04-20
WO 03/059251
PCT/US02/33991
0 0
0
0
Nzz.? y0
0, NH H N ¨CO2H
NH
0
Targeting Agent Linker with 1,3
Diketone Group
SCS864
[0087] A linker reactive
group chemical moiety (Z )suitable for covalent
modification by a reactive sulfhydryl group in an antibody may be a disulfide,
aryl
halide, maleimide, alpha-haloacetamide, isocyanate, epoxide, thioester, active
ester,
amidine, guanidine, imine, eneamine, phosphate, phosphonate, epoxide,
aziridine,
thioepoxide, masked or protected diketone (ketal for example), lactam,
haloketone,
aldehyde, and the like. The chemical structures of various targeting agent-
linker
compounds which include a linker with a 1,3 diketone as the reactive group are
shown
in FIGs. 2-5.
[0088] One of skill in the art will readily appreciate that reactive amino
acid side
chains in antibodies may possess an electrophilic group that reacts with a
nucleophilic
group on the targeting agent or its linker, whereas in other embodiments a
reactive
nucleophilic group in an amino acid side chain of a combining site of an
antibody or an
antibody fragment reacts with an electrophilic group in a targeting agent or
linker.
Thus, antibody or antibody fragment combining site side chains may be
substituted
with an electrophile (e.g., FIGs. 6 and 7 ) and this group may be used to
react with a
nucleophile on the targeting agent or its linker (e.g., NI-12). In this
embodiment, the
antibody and targeting agent each have a partial linker with appropriate
reactive
moieties at each end so that the two ends of the partial linker can form the
full linker,
thus creating the complete targeting compound.
33

CA 02464472 2004-04-20
WO 03/059251
PCT/US02/33991
[0089] One of skill in the art also will readily appreciate that two or
more targeting
agents may be linked to a single antibody combining site. The two targeting
agents may
be the same or may be different with respect to their specificity for a
particular target.
In one embodiment, each targeting agent may be linked to a separate reactive
side chain
of an amino acid in the antibody combining site. In a preferred embodiment,
the two
targeting agents are attached to a branched or linear linker which then links
both
targeting agents to the same reactive amino acid side chain in the antibody
combining
site. Each branch of a branched linker may in some embodiments comprise a
linear
stretch of between 5-100 atoms. By way of example, the structures disclosed in
FIGs.
3-5 show embodiments of branched linkers with two targeting agents linked to a

different branch of the linker, which has a 1,3-diketone as the reactive
group. As
shown in these embodiments, the branch point may be in the connecting chain or
in the
recognition group (if present).
[0090] "Antibody" as used herein includes immunoglobulins which are the
product
of B cells and variants thereof as well as the T cell receptor (TcR) which is
the product
of T cells and variants thereof. An immunoglobulin is a protein comprising one
or
more polypeptides substantially encoded by the immunoglobulin kappa and
lambda,
alpha, gamma, delta, epsilon and mu constant region genes, as well as myriad
immunoglobulin variable region genes. Light chains are classified as either
kappa or
lambda. Heavy chains are classified as gamma, mu, alpha, delta, or epsilon,
which in
turn define the immunoglobulin classes, IgG, IgM, IgA, IgD and IgE,
respectively.
Also subclasses of the heavy chain are known. For example, IgG heavy chains in

humans can be any of IgG 1 , IgG2, IgG3 and IgG4 subclass.
[0091] A typical immunoglobulin structural unit is known to comprise a
tetramer.
Each tetramer is composed of two identical pairs of polypeptide chains, each
pair
having one "light" (about 25 IcD) and one "heavy" chain (about 50-70 IcD). The
N-
terminus of each chain defines a variable region of about 100 to 110 or more
amino
acids primarily responsible for antigen recognition. The terms variable light
chain (VL)
and variable heavy chain (VH) refer to these light and heavy chains
respectively.
34

CA 02464472 2004-04-20
WO 03/059251
PCT/US02/33991
[0092] Antibodies exist as full length intact antibodies or as a number of
well-
characterized fragments produced by digestion with various peptidases or
chemicals.
Thus, for example, pepsin digests an antibody below the disulfide linkages in
the hinge
region to produce F(ab')2, a dimer of Fab which itself is a light chain joined
to VH-CHI
by a disulfide bond. The F(ab')2 may be reduced under mild conditions to break
the
disulfide linkage in the hinge region thereby converting the F(ab')2 dimer
into an Fab'
monomer. The Fab' monomer is essentially a Fab fragment with part of the hinge

region (see, Fundamental Immunology, W. E. Paul, ed., Raven Press, N.Y.
(1993), for a
more detailed description of other antibody fragments). While various antibody

fragments are defined in terms of the digestion of an intact antibody, one of
skill will
appreciate that any of a variety of antibody fragments may be synthesized de
novo
either chemically or by utilizing recombinant DNA methodology. Thus, the term
antibody, as used herein also includes antibody fragments either produced by
the
modification of whole antibodies or synthesized de novo or antibodies and
fragments
obtained by using recombinant DNA methodologies.
[0093] The T cell receptor (TcR) is a disulfide linked heterodimer composed
of a
or 13 chains or, on a minority of T cells, y or 8 chains. The two chains are
generally
disulfide-bonded just outside the T cell plasma membrane in a short extended
stretch of
amino acids resembling the antibody hinge region. Each TcR chain is composed
of one
Antibody-like variable domain (Va or Vf3) and one constant domain (Ca or Cu).
The
full TcR has a molecular mass of about 95 kDa with the individual chains
varying in
size from 35 to 47 IcDa. Also encompassed within the meaning of TCR are
portions of
the receptor such as the variable regions of this receptor that can be
produced as a
soluble protein using methods well known in the art. For example, U.S. Patent
No.
6,080,840 describes a soluble T cell receptor (TcR) prepared by splicing the
extracellular domains of a TcR to the glycosyl phosphatidylinositol (GPI)
membrane
anchor sequences of Thy-1. The molecule is expressed in the absence of CD3 on
the
cell surface, and can be cleaved from the membrane by treatment with
phosphatidylinositol specific phospholipase C (PI-PLC). The soluble TcR also
may be
prepared by coupling the TcR variable domains to an antibody heavy chain CH2
or CH3

CA 02464472 2004-04-20
WO 03/059251
PCT/US02/33991
domain, essentially as described in U.S. Patent No. 5,216,132 or as soluble
TcR single
chains as described by Schusta et al. Nature Biotech. 18,754-759 (2000) or
Holler et al.
Proc. Natl. Acad. Sci (USA) 97:5387-5392 (2000). The TcR "antibodies" as
soluble
products may be used in place of antibody for making the compounds of the
invention.
The combining site of the TcR can be identified by reference to CDR regions
and other
framework residues using the same methods discussed above for antibodies.
[0094] Recombinant antibodies may be conventional full length antibodies,
antibody fragments known from proteolytic digestion, unique antibody fragments
such
as Fv or single chain Fv (scFv), domain deleted antibodies, and the like. An
Fv
antibody is about 50 Kd in size and comprises the variable regions of the
light and
heavy chain. A single chain Fv ("scFv") polypeptide is a covalently linked
VH::VL
heterodimer which may be expressed from a nucleic acid including VH- and VL-
encoding sequences either joined directly or joined by a peptide-encoding
linker. See
Huston, et al. (1988) Proc. Nat. Acad. Sci. USA, 85:5879-5883. A number of
structures
for converting the naturally aggregated, but chemically separated light and
heavy
polypeptide chains from an antibody V region into an scFv molecule which will
fold
into a three dimensional structure substantially similar to the structure of
an antigen-
binding site. See, e.g. U.S. Patent Nos. 5,091,513, 5,132,405 and 4,956,778.
[0095] The combining site refers to the part of an antibody molecule that
participates in antigen binding. The antigen binding site is formed by amino
acid
residues of the N-terminal variable ("V") regions of the heavy ("H") and light
("L")
chains. The antibody variable regions comprise three highly divergent
stretches
referred to as "hypervariable regions" or "complementarity determining
regions"
(CDRs) which are interposed between more conserved flanking stretches known as

"framework regions" (FRs). In an antibody molecule, the three hypervariable
regions
of a light chain (LCDR1, LCDR2, and LCDR3) and the three hypervariable regions
of a
heavy chain (HCDR1, HCDR2 and HCDR3) are disposed relative to each other in
three
dimensional space to form an antigen binding surface or pocket. The antibody
36

CA 02464472 2004-04-20
WO 03/059251
PCT/US02/33991
combining site therefore represents the amino acids that make up the CDRs of
an
antibody and any framework residues that make up the binding site pocket.
[0096] The identity of the amino acid residues in a particular antibody
that make
up the combining site can be determined using methods well known in the art.
For
example, antibody CDRs may be identified as the hypervariable regions
originally
defined by Kabat et al. (see, "Sequences of Proteins of Immunological
Interest," E.
Kabat et al., U.S. Department of Health and Human Services; Johnson, G and Wu,
TT
(2001) Kabat Database and its applications: future directions. Nucleic Acids
Research,
29: 205-206; http://immuno.bme.nwa.edu). The positions of the CDRs may also be

identified as the structural loop structures originally described by Chothia
and others,
(see Chothia and Lesk, J. Mol. Biol. 196, 901 (1987), Chothia et al., Nature
342, 877
(1989), and Tramontano et al., J. Mol. Biol. 215, 175 (1990)). Other methods
include
the "AbM definition" which is a compromise between Kabat and Chothia and is
derived using Oxford Molecular's AbM antibody modeling software (now Accelrys)
or
the "contact definition" of CDRs by Macallum et al., ("Antibody-antigen
interactions:
contact analysis and binding site topography," J Mol Biol. 1996 Oct
11;262(5):732-45).
The following chart identifies CDRs based upon various known definitions.
Loop Kabat AbM Chothia Contact
Li L24 -- L34 L24 -- L34 L24 -- L34 L30 -- L36
L2 L50 -- L56 L50 -- L56 L50 -- L56 L46 -- L55
L3 L89 -- L97 L89 -- L97 L89 -- L97 L89 -- L96
H1 H31 -- H35B H26 -- H35B H26 -- H32..34 H30 -- H35B
(Kabat Numbering)
H1 H31 -- H35 H26 -- H35 H26 -- H32 H30 -- H35
(Chothia Numbering)
H2 H50 -- H65 H50 -- H58 H52 -- H56 H47 -- H58
H3 H95 -- H102 H95 -- H102 H95 -- H102 H93 -- H101
37

CA 02464472 2004-04-20
WO 03/059251
PCT/US02/33991
General guidelines by which one may identify the CDRs in an antibody from
sequence
alone are as follows:
LCDR1:
Start - Approximately residue 24.
Residue before is always a Cys.
Residue after is always a Trp. Typically TRP is followed with TYR-GLN, but
also
may be followed by LEU-GLN, PHE-GLN, or TYR-LEU.
Length is 10 to 17 residues.
LCDR2:
Start - 16 residues after the end of Li.
Sequence before is generally ILE-TYR, but also may be VAL-TYR, ILE-LYS, or ILE-

PHE.
Length is generally 7 residues.
LCDR3:
Start ¨ generally 33 residues after end of L2.
Residue before is a Cys.
Sequence after is PHE-GLY-X-GLY.
Length is 7 to 11 residues.
HCDR1:
Start ¨ at approximately residue 26 (four residues after a CYS) [Chothia / AbM

definition] Kabat definition starts 5 residues later.
Sequence before is CYS-X-X-X.
Residues after is a TRP, typically followed by VAL, but also followed by ILE,
or
ALA.
Length is 10 to 12 residues under AbM definition while Chothia definition
excludes the
last 4 residues.
38

CA 02464472 2004-04-20
WO 03/059251
PCT/US02/33991
HCDR2:
Start - 15 residues after the end of Kabat /AbM definition of CDR-H1.
Sequence before typically LEU-GLU-TRP-ILE-GLY (SEQ ID NO. 1), but a number of
variations are possible.
Sequence after is LYS/ARG-LEU/ILENAL/PHE/THR/ALA-THR/SER/ILE/ALA
Length is 16 to 19 residues under Kabat definition (AbM definition ends 7
residues
earlier).
HCDR3:
Start ¨33 residues after end of CDR-H2 (two residues after a CYS).
Sequence before is CYS-X-X (typically CYS-ALA-ARG).
Sequence after is TRP-GLY-X-GLY.
Length is 3 to 25 residues.
[0097] The identity of the amino acid residues in a particular antibody
that are
outside the CDRs, but nonetheless make up part of the combining site by having
a side
chain that is part of the lining of the combining site (i.e., it is available
to linkage
through the combining site), can be determined using methods well known in the
art
such as molecular modeling and X-ray crystallography. See e.g., Riechmann et
al.,
(1988) Nature, 332:;323-327. The aldolase antibody mouse mAb 38C2, which has a

reactive lysine near to but outside HCDR3, is an example of such an antibody.
[0098] The reactive residue of the antibody combining site may be naturally
associated with the antibody such as when the residue is encoded by nucleic
acid
present in the lymphoid cell first identified to make the antibody.
Alternatively, the
amino acid residue may arise by purposely mutating so as to encode the
particular
residue (see, e.g., WO 01/22922 to Meares et al.). In another approach, the
amino acid
residue or its reactive elements (e.g., a nucleophilic amino group or
sulfhydryl group)
may be attached to an amino acid residue in the antibody combining site. Thus,

covalent linkage with the antibody occurring "through an amino acid residue in
the
combining site of the antibody" as used herein means that linkage can be
directly to an
39

CA 02464472 2004-04-20
WO 03/059251
PCT/US02/33991
amino acid residue of an antibody combining site or through a chemical moiety
that is
linked to a side chain of an amino acid residue of an antibody combining site.
100991 As discussed, antibodies that can be used in preparing the antibody
targeting compounds of the invention require a reactive side chain in the
antibody
combining site. A reactive side chain may be present or be placed by mutation
in any
antibody. Catalytic antibodies are a preferred source of such antibodies. Such

antibodies include aldolase antibodies, beta lactamase antibodies, esterase
antibodies,
amidase antibodies, and the like.
[00100] A reactive lysine in an antibody combining site may be covalently
linked to
a ketone, diketone, beta lactam, active ester haloketone, lactone, anhydride,
maleimide,
epoxide, aldehyde amidine, guanidine, imines, eneamines, phosphates,
phosphonates,
epoxides, aziridines, thioepoxides, masked or protected diketones (ketals for
example),
lactams, haloketones, aldehydes, and the like, associated with a targeting
agent or
linker-targeting agent. An exemplary and preferred such antibody is an
aldolase
antibody such as the mouse monoclonal antibody mAb 38C2 and other like
catalytic
antibodies as well as suitably humanized and chimeric versions of such
antibodies.
Mouse mAb 38C2 is the prototype of a new class of catalytic antibodies that
were
generated by reactive immunization and mechanistically mimic natural aldolase
enzymes (Barbas et al., 1997, Science 278, 2085-2092). Through a reactive
lysine,
these antibodies catalyze aldol and retro-aldol reactions using the enamine
mechanism
of natural aldolases (Wagner et al., 1995, Science 270, 1797-1800; Barbas et
al., 1997,
Science 278, 2085-2092; Zhong et al., 1999, Angew. Chem. Int. Ed. 38, 3738-
3741;
Karlstrom et al., 2000, Proc. Natl. Acad. Sci. U.S.A., 973878-3883). In
addition to
their versatility and efficacy in synthetic organic chemistry (e.g., Hoffmann
et al., 1998,
J. Am. Chem. Soc. 120, 2768-2779 ; Sinha et al., 1998, Proc. Natl. Acad. Sci.
U.S.A.
95, 14603-14608 ), aldolase antibodies have been used to activate
camptothecin,
doxorubicin, and etoposide prodrugs in vitro and in vivo as an anti-cancer
strategy
(Shabat et al., 1999, Proc. Natl. Acad. Sci. U.S.A. 96, 6925-6930 and ,2001,
Proc. Natl.
Acad. Sci. U.S.A. 98, 7528-7533).

CA 02464472 2004-04-20
WO 03/059251
PCT/US02/33991
[00101] In another example, the reactive amino acid of an antibody
combining site
may be a reactive cysteine, serine or tyrosine residue. For cysteines, the
resulting
antibody may form a covalent linkage with maleimide-containing components or
other
thiol-reactive groups such as iodoacetamides, aryl halides, disulfhydryls and
the like.
Reactive cysteines may be found in thioesterase catalytic antibodies as
described by
Janda et al., Proc. Natl. Acad. Sci. (USA) 91:2532-2536, (1994). For other
esterase
antibodies see Wirsching et al., Science 270:1775-82 (1995). Reactive amino
acid-
containing antibodies may be prepared by means well known in the art including

mutating an antibody combining site residue to encode for the reactive amino
acid or
chemically derivatizing an amino acid side chain in an antibody combining site
with a
linker that contains the reactive group.
[00102] Antibodies suitable for use herein may be obtained by conventional
immunization, reactive immunization in vivo, or by reactive selection in
vitro, such as
with phage display. Antibodies may be produced in humans or in other animal
species.
Antibodies from one species of animal may be modified to reflect another
species of
animal. For example, human chimeric antibodies are those in which at least one
region
of the antibody is from a human immunoglobulin. A human chimeric antibody is
typically understood to have variable regions from a non-human animal, e.g. a
rodent,
with the constant regions from a human. In contrast, a humanized antibody uses
CDRs
from the non-human antibody with most or all of the variable framework regions
from
and all the constant regions from a human immunoglobulin. Chimeric and
humanized
antibodies may be prepared by methods well known in the art including CDR
grafting
approaches (see, e.g., U.S. Patent Nos. 5,843,708; 6,180,370; 5,693,762;
5,585,089;
5,530,101), chain shuffling strategies (see e.g., U.S. Patent No. 5,565,332;
Rader et al.,
Proc. Natl. Acad. Sci. USA (1998) 95:8910-8915), molecular modeling strategies
(U.S.
Patent No. 5,639,641), and the like.
[00103] Unlike typical chemical derivatization of antibodies, those derived
from
reactive immunization can be specifically labeled in their binding site at a
defined
position, facilitating the rapid and controlled preparation of a homogeneous
product. In
41

CA 02464472 2004-04-20
WO 03/059251
PCT/US02/33991
addition, unlike chemical derivatization of antibodies, those derived from
reactive
immunization with 1,3-diketones are reversible. Due to this reversibility, a
diketone
derivative of a targeting compound bound to mAb 38C2 can be released from the
antibody through competition with the covalent binding hapten JW (Wagner et
al.,
1995, Science 270, 1797-800), or related compounds. This allows one to
immediately
neutralize the conjugate in vivo in case of an adverse reaction.
Alternatively, non-
reversible covalent linkage is possible such as with aldolase antibodies and
beta lactam
derivatives of the targeting compound. Unlike typical anti-hapten antibodies,
covalent
diketone binding antibodies have the advantage that the covalent linkage that
is formed
between the diketone and antibody is stable to large changes in pH, either
extremes of
low pH 3 or high pH 11. Such pH shifts do not release the targeting compound
from
the antibody. This is an advantage for tumor targeting since tumors typically
exhibit
reduced pH as compared to normal tissues. The added stability of covalent
binding
antibodies covalently linked to their targeting agent should provide
additional
advantages in terms of formulation, delivery, and long term storage.
[00104] A targeting compound of the present invention can be made using
techniques well known in the art. Typically, synthesis of a targeting agent
which also is
a functional component (biological agent) is the first step. The targeting
agent (also
functional component in this case) is then derivatized for linkage to a
connecting
component (the linker) which is then combined with the antibody. One of skill
in the
art will readily appreciate that the specific synthetic steps used depend upon
the exact
nature of the three components.
[00105] By way of example, as a first step, targeting agent-linker
compounds shown
as Compounds 15 and 4, was made as shown in Schemes 1 (FIG. 10) and 2 (FIG.
11),
respectively, as derivatized versions of the integrin targeting agents shown
as
Compounds 1 and 2, above. Compounds 15 and 4 were derivatized (relative to
Compounds 1 and 2) by addition of a portion of the linker (connecting
component).
Scheme 3 (FIG. 12) shows additional synthetic steps by which a complete linker
with a
42

CA 02464472 2004-04-20
WO 03/059251
PCT/US02/33991
diketone reactive moiety was added to derivatized targeting agent Compound 15
to
obtain targeting compounds SCS-873 and SCS-1655.
[00106] Integrin targeting components shown as compounds 15 and 4 were
synthesized as shown in the FIG. 10 (Scheme 1) and FIG. 11 (Scheme 2),
respectively.
A linker with a diketone reactive moiety was added to these targeting
molecules as
shown in Scheme 3 (FIG. 12) to form targeting compound-linker molecules SCS-
873
and SCS-1655. Synthesis of SCS-873 was achieved starting from compound 14 in
three steps. Compound 14 was converted to 15 as shown in Scheme 1 and the
crude
product was reacted with an N-hydroxysuccinimide (NHS) -ester of the diketone
compound 23 in CH3CN-DMF in the presence of Et3N. Purification over silica gel

(CH2C12-Me0H, 9:1) afforded pure SCS-873.
[00107] Compound SCS-1655 was synthesized from 14 in five steps (Schemes 2
and 3). Deprotection of the BOC group in compound 14 followed by reaction with
the
NHS ester of the bivalent linker 24 afforded compound 25, which was then
deprotected
and reacted with 23 as above to afford SCS-1655.
[00108] Synthesis of integrin targeting component-linker molecules SCS-864
and
SCS-789 is shown in Scheme 4 (FIG. 13). SCS-864 and SCS-789 were each
synthesized in one step from compound 4 (FIG. 13, scheme 4). Linking of
Compound
4 was achieved with the appropriate activated NHS-ester.
[00109] Targeting agent-linker compounds, such as SCS-864, SCS-873 and SCS-
1655 where the linker includes a diketone reactive moiety, can be incubated
with 0.5
equiv. of an aldolase antibody such as mAb 38C2 to produce antibody targeting
compounds. Additional examples are set forth below.
[00110] Also provided are targeting agent-linker compounds for covalently
linking
to a combining site of an antibody. The linker is of sufficient length to
allow the
targeting agent to bind to the target molecule when the targeting agent is
linked through
the linker to an antibody. In some embodiments, the targeting agent-linker
compound
43

CA 02464472 2004-04-20
WO 03/059251
PCT/US02/33991
includes one or more targeting agents specific for a target molecule with a
linker of the
formula X ¨ Y ¨ Z. The makeup of linker components X, Y and Z are as described

above. If two or more targeting agents are included in the targeting agent-
linker
compound, the various targeting agents may be attached directly to the linker
or the
linker may be branched with targeting agents attached to different linker
branches.
1001111 Also provided is a targeting agent-linker compound that can be
noncovalently associated with the combining site of an antibody. This compound
can
be used in conjunction with a suitable antibody to form a targeting compound
of the
invention. Such targeting agent-linker compounds comprise two or more
targeting
agents covalently linked via a linker to an antigen recognized by the
antibody. The
linker may linear or branched and should be of sufficient length to allow the
targeting
agent(s) to bind to the target molecule when the targeting agent(s) is linked
through the
linker to the antibody.
[00112] In some embodiments, the linker includes any of C, H, N, 0, P, S,
Si, F, Cl,
Br, and I, or a salt thereof. The linker also may include a group such as an
alkyl,
alkenyl, alkynyl, oxoalkyl, oxoalkenyl, oxoalkynyl, aminoalkyl, aminoalkenyl,
aminoalkynyl, sulfoalkyl, sulfoalkenyl, or sulfoalkynyl group, phosphoalkyl,
phosphoalkenyl, phosphoalkynyl group. The linker also may include one or more
ring
structures. Combinations of the above groups and rings may also be present in
the
linkers of the targeting compounds of the invention. In some embodiments, the
linker
has a linear stretch of between 2-200 atoms although in other embodiments,
longer
linker lengths may be used. One or more targeting agents may be linked to the
linker
and if a branched linker is used, some of the targeting agents may be linked
to different
branches of the linker.
[00113] In some embodiments, the targeting agent of the targeting agent-
linker
compound is biologically active while in other embodiments, the targeting
agent-linker
compound further includes a separate biological agent, which is preferably
covalently
linked to the targeting agent. In some embodiments, the biological agent may
be linked
44

CA 02464472 2004-04-20
WO 03/059251
PCT/US02/33991
to the targeting agent or to the linker using essentially the same approaches
used to link
the targeting agent to the linker or using other approaches well known in the
art.
[00114] The antigen of the linker can be any antigen which can be bound by
an
available antibody. Antigens are well known in the art and include, an organic

compound, a drug, a biomolecule such as a protein, peptide, peptidomimetic,
glycoprotein, proteoglycan, lipid, glycolipid, nucleic acid, carbohydrates,
and the like
as well as combinations of these molecules.
[00115] The present invention also includes methods of modifying the
combining
site of an antibody to generate binding specificity for a particular target
molecule. Such
methods include covalently linking a reactive amino acid side chain in the
combining
site of the antibody to a chemical moiety on a linker of a targeting agent-
linker
compound where the targeting agent is specific for the target molecule. The
chemical
moiety of the linker is sufficiently distanced from the targeting agent so
that the
targeting agent can bind to the target molecule when the targeting agent-
linker
compound is covalently linked to the antibody combining site. Typically, the
antibody
will not be considered specific for the target molecule. In a preferred
embodiment, the
antibody prior to covalent linking would have an affinity for the target
molecule of less
than about 1 x 10-5 moles/liter. However, after the antibody is covalently
linked to the
targeting agent-linker compound, the modified antibody preferably has an
affinity for
the target molecule of at least about 1 x 10-6 moles/liter, more preferably at
least about
1 x 10-7 moles/liter, even more preferably at least 1 x 10-8 moles/liter, yet
even more
preferably at least 1 x 10-9 moles/liter, most preferably at least about 1 x
10-'9
moles/liter.
[001161 The present invention also includes methods of altering at least
one
physical or biological characteristic of a targeting agent, biological agent
or linker. The
methods include covalently linking the targeting agent or biological agent to
the
combining site of an antibody as described above. In some embodiments, the
targeting
agent or biological agent is linked to the antibody combining site though a
linker, the
characteristics of which are described above. The method is particularly
useful for

CA 02464472 2004-04-20
WO 03/059251
PCT/US02/33991
linking small targeting or biological agents of 5 Kd or less. However, the
method also
works for larger such molecules. Characteristics of the targeting agent or
biological
agent can include binding affinity, susceptibility to degradation, such as by
proteases,
pharmocokinetics, pharmacodynamics, immunogenicity, solubility, solubility,
lipophilicity, hydrophilicity, hydrophobicity, stability (more or less stable
as well as
planned degradation), rigidity, flexibility, modulation of antibody binding,
and the like.
[00117] As used herein, pharmacokinetics refers to the concentration an
administered compound in the serum over time. Pharmacodynamics refers to the
concentration of an administered compound in target and nontarget tissues over
time
and the effects on the target tissue (efficacy) and the non-target tissue
(toxicity).
Improvements in, for example, pharmacokinetics or pharmacodynamics can be
designed for a particular targeting agent or biological agent such as by using
labile
linkages or by modifying the chemical nature of any linker (changing
solubility, charge,
etc.).
[00118] The biological characteristic of an antibody targeting compound of
the
invention may be modified to obtain improved pharmaceutical or other
characteristics.
This may be achieved by altering one or more chemical characteristics of the
targeting
agent or biological agent, the linker or the antibody. A preferred approach is
to
chemically modify one or more chemical characteristics of the linker. By
altering
chemical characteristics of the compound including the linker, one can obtain
improved
features such as improvement in pharmockinetics, pharmacodynamics, solubility,

immunogenicity and the like.
[00119] The targeting compounds of the present invention have many uses.
For
example, the antibody portion of a targeting compound may generally extend the
half-
life of a smaller sized targeting agent in vivo. Also, the biological potency
of a
particular targeting agent may be increased by the addition of effector
function(s)
provided by the antibody portion of the targeting compound (e.g., complement
mediated effector functions). In addition, the targeting agent, through its
increased size
conferred by linkage to the antibody, may enable the targeting agent to
function as a
46

CA 02464472 2004-04-20
WO 03/059251
PCT/US02/33991
competitive inhibitor in situations where it would otherwise fail to do so.
Thus, in one
aspect, the invention provides a method for increasing the effective
circulating half-life
of a targeting agent. The method includes linking the targeting agent to an
antibody
using a linking group as set forth above. In another aspect, the invention
provides a
method of redirecting an antibody to a specific target. The method includes
linking an
antibody to a targeting agent through a linker as set forth above.
1001201 The invention also provides a method of treating or preventing a
disease or
condition in an individual wherein said disease or condition involves cells,
tissue or
fluid that expresses a target molecule. The method includes administering to a
subject
such as a patient, a therapeutically effective amount of a targeting compound
of the
invention. The subject may be an animal such as a mammal. In some embodiments,

the subject is a human. The compound may include a biological agent that is
the same
or is distinct from the targeting agent and which may take any of the forms or
activities
described herein. In some preferred embodiments, the target molecule is an
integrin
and the disease is a carcinoma. The association of integrin expression in
carcinomas is
well known in the art (See, e.g., United States Patent Nos. 5,753,230 and
5,766,591, the
disclosures of which are incorporated herein by reference). For therapeutic
use in
humans, a human, humanized, or human chimeric antibody is a preferred as the
antibody component of the targeting compound. An antibody with a human IgG4
constant region also is preferred if agonist activity is desired.
[00121] In addition to therapeutic applications, antibody targeting
compounds of the
invention may also be used for the imaging of cells such as tumor cells or
tissues (e.g.,
an extracellular matrix biomolecule) as is well known in the art. Accordingly,
provided
is a method of imaging cells or tissue (e.g., an extracellular matrix
biomolecule) in an
individual. In such methods, the cells or tissue expresses a target molecule.
The
method includes administering to a subject an antibody targeting compound of
the
invention linked to a detectable label. A detectable label for use in such
methods can
be a radioisotope or may be a non-radioisotope such as may be used in nuclear
magnetic resonance (NMR) imaging. In the latter case, one may link the
antibody
47

CA 02464472 2004-04-20
WO 03/059251
PCT/US02/33991
targeting agent to chelates e.g., diethylenetriaminepentaacetate (DTPA) of the

paramagnetic metal gadolinium essentially as described in Simkins et al., Nat.
Med.,
4(5):623-6 (1998).
[00122] The binding of a mixture of SCS-873 and 38C2 to human Karposi's
sarcoma SLK cells was studied. SCS-873 effectively mediated cell surface
binding of
38C2. No binding of 38C2 was detectable in the absence of SCS-873. Control
experiments confirmed that the 1,3- diketone moiety is required for binding of
SCS-873
to 38C2. After independent i.p. and i.v. injections, respectively, SCS-873 and
38C2
form an integrin avi33 targeting conjugate in vivo. In these experiments, the
circulatory
half-life of SCS-873 was extended by more than two orders of magnitude through

binding to 38C2. Combination of SCS-873 and 38C2 effectively inhibited tumor
growth in a mouse model of human Karposi's sarcoma, whereas either SCS-873 or
38C2 alone were less effective or not effective at all.
[00123] The present invention also provides methods of targeting a
biological
activity to cells, tissue (e.g., an extracellular matrix biomolecule) or a
biolomolecule in
the fluid of a subject. The method includes administering to the subject, a
targeting
compound that includes a targeting agent specific for the cells, tissue
extracellular
matrix biomolecule or fluid biomolecule. The targeting agent is covalently
linked to an
amino acid residue in the combining site of an antibody. In some embodiments,
a
linker is used to link the targeting agent to the antibody. The targeting
agent is not an
antibody. In some embodiments, the compound has a biological activity while in
other
embodiments, an biologically active molecule that is not the targeting agent
is included
as a component of the compound. Alternatively, the component parts of the
targeting
compound may be separately administered and then form the covalent compound in

vivo. In such a method, the targeting agent may include a linker/reactive
moiety or the
antibody combining site may be suitably modified to covalently link to the
targeting
agent.
48

CA 02464472 2004-04-20
WO 03/059251
PCT/US02/33991
[00124] A targeting compound of the present invention can be administered
as a
pharmaceutical or medicament that includes a targeting compound of the
invention
formulated with a pharmaceutically acceptable carrier. Accordingly, the
compounds
may be used in the manufacture of a medicament or pharmaceutical composition.
Pharmaceutical compositions of the invention may be formulated as solutions or

lyophilized powders for parenteral administration. Powders may be
reconstituted by
addition of a suitable diluent or other pharmaceutically acceptable carrier
prior to use.
Liquid formulations may be buffered, isotonic, aqueous solutions. Powders also
may
be sprayed in dry form. Examples of suitable diluents are normal isotonic
saline
solution, standard 5% dextrose in water, or buffered sodium or ammonium
acetate
solution. Such formulations are especially suitable for parenteral
administration, but
may also be used for oral administration or contained in a metered dose
inhaler or
nebulizer for insufflation. It may be desirable to add excipients such as
polyvinylpyrrolidone, gelatin, hydroxy cellulose, acacia, polyethylene glycol,
mannitol,
sodium chloride, sodium citrate, and the like.
[00125] Alternately, compounds may be encapsulated, tableted or prepared in
an
emulsion or syrup for oral administration. Pharmaceutically acceptable solid
or liquid
carriers may be added to enhance or stabilize the composition, or to
facilitate
preparation of the composition. Solid carriers include starch, lactose,
calcium sulfate
dihydrate, terra alba, magnesium stearate or stearic acid, talc, pectin,
acacia, agar or
gelatin. Liquid carriers include syrup, peanut oil, olive oil, saline and
water. The
carrier may also include a sustained release material such as glyceryl
monostearate or
glyceryl distearate, alone or with a wax. The amount of solid carrier varies
but,
preferably, will be between about 20 mg to about 1 g per dosage unit. The
pharmaceutical preparations are made following the conventional techniques of
pharmacy involving milling, mixing, granulating, and compressing, when
necessary, for
tablet forms; or milling, mixing and filling for hard gelatin capsule forms.
When a
liquid carrier is used, the preparation may be in the form of a syrup, elixir,
emulsion, or
an aqueous or non-aqueous suspension. For rectal administration, the invention
49

CA 02464472 2004-04-20
WO 03/059251
PCT/US02/33991
compounds may be combined with excipients such as cocoa butter, glycerin,
gelatin or
polyethylene glycols and molded into a suppository.
[00126] Compounds of the invention may be formulated to include other
medically
useful drugs or biological agents. The compounds also may be administered in
conjunction with the administration of other drugs or biological agents useful
for the
disease or condition that the invention compounds are directed.
[00127] As employed herein, the phrase "an effective amount," refers to a
dose
sufficient to provide concentrations high enough to impart a beneficial effect
on the
recipient thereof. The specific therapeutically effective dose level for any
particular
subject will depend upon a variety of factors including the disorder being
treated, the
severity of the disorder, the activity of the specific compound, the route of
administration, the rate of clearance of the compound, the duration of
treatment, the
drugs used in combination or coincident with the compound, the age, body
weight, sex,
diet, and general health of the subject, and like factors well known in the
medical arts
and sciences. Various general considerations taken into account in determining
the
"therapeutically effective amount" are known to those of skill in the art and
are
described, e.g., in Gilman et al., eds., Goodman And Gilman's: The
Pharmacological
Bases of Therapeutics, 8th ed., Pergamon Press, 1990; and Remington's
Pharmaceutical
Sciences, 17th ed., Mack Publishing Co., Easton, Pa., 1990. Dosage levels
typically
fall in the range of about 0.001 up to 100 mg/kg/day; with levels in the range
of about
0.05 up to 10 mg/kg/day are generally applicable. A compound can be
administered
parenterally, such as intravascularly, intravenously, intraarterially,
intramuscularly,
subcutaneously, or the like. Administration can also be orally, nasally,
rectally,
transdermally or inhalationally via an aerosol. The composition may be
administered
as a bolus, or slowly infused.
[00128] The administration of an antibody-targeting agent conjugate to an
immunocompetent individual may result in the production of antibodies against
the
conjugate. Such antibodies may be directed to the antibody itself, such as the
variable
region including the antibody idiotype as well as to the targeting agent or
any linker

CA 02464472 2004-04-20
WO 03/059251
PCT/US02/33991
used to conjugate the targeting agent to the antibody. Reducing the
immunogenicity of
the antibody-targeting agent conjugate can be addressed by methods well known
in the
art such as by attaching long chain polyethylene glycol (PEG)-based spacers,
and the
like, to the antibody-targeting agent. Long chain PEG and other polymers are
known
for their ability to mask foreign epitopes, resulting in the reduced
immunogenicity of
therapeutic proteins that display foreign epitopes (Katre et al., 1990,1
Immunol. 144,
209-213; Francis et al., 1998, Int. J. Hematol. 68, 1-18). As noted, PEG can
be a linker
as well, thus providing both linker function and reduced immunogenicity in a
targeting
compound of the invention. Alternatively, or in addition, the individual
administered
the antibody-targeting agent conjugate may be administered an
immunosuppressent
such as cyclosporin A, anti-CD3 antibody, and the like.
1001291 A method of screening a chemical library for agonists or
antagonists of a
receptor is further provided. The method includes linking individual members
of the
chemical library to the combining site of an antibody and then testing the
antibody
linked library for binding to the receptor or for inhibition of binding
between the
receptor and a ligand for the receptor. By this approach, the present antibody
targeting
compounds provide a new format for high throughput screening to identify
candidate
small molecule chemicals such as drugs peptides peptidomimetics, organic
compounds,
and the like, that function for example, as antagonists or agonists. The
relative small
size of a useful candidate chemical molecule typically requires indirect
screening such
as in displacement or competition formats. As provided herein, one can build
the
chemical library on an antibody format, by linking individual drugs in the
library to a
combining site of an antibody.
1001301 Antibody combining site-tagged libraries may be prepared by
synthesizing
chemical candidates with a suitable linker comprising a particular linker
moiety
designed for covalent interaction with a particular antibody. Such linkers may
include a
diketone moiety to be used in conjunction with an aldolase antibody that
includes a
reactive lysine in the combining site. One skilled in the art would readily
understand
51

CA 02464472 2004-04-20
WO 03/059251
PCT/US02/33991
that other linkers and linker moieties (e.g., biotin) which have been
described herein are
clearly useful for this purpose.
[00131] Antibody combining site-tagged chemical libraries thus prepared can
be
used, for example, in receptor assays or cell bioassays where binding of each
compound
in the library may be monitored by detecting the linked antibody. Detection of
the
antibody portion of each compound may be accomplished by methods of antibody
detection well known in the art. For example, the antibody may be linked to a
detectable moiety such as an enzyme, fluorophore, radioisotope, and the like.
Indirect
systems can also be used such as biotin-streptavidin. Libraries can be
screened on cells
or impure antigens such as viral lysates as well as on purified antigens. For
example,
libraries can be tested for binding or inhibition of binding using as the
target, lysates
run on protein gels, with the analysis focussed on a particular gel band. In
the case
where the receptor is expressed on a cell, binding or inhibition of binding
can be
determined by detecting cellular signaling events occurring (or not occurring
as in the
case of inhibition) downstream of said binding or inhibition of binding.
Downstream
cellular signaling can be detected with the aid of a reporter gene as is well
known in the
art (see, e.g., U.S. Patent No. 5,618,720 and 5,670,113).
[00132] Screening of antibody tagged chemical libraries can be readily
adapted for
use with high throughput instruments. Screening may be done in vitro or in
vivo.
Furthermore, a biological display library such as a peptide phage library may
be used to
prepare an antibody combining site-tagged library. In such cases, the site of
attachment
of the linker moiety (e.g., diketone) can be the fusion point of the library
to the
biological carrier.
[00133] Also provided is an immunoassay method for determining the amount
of
analyte in a sample. Such methods include:
(a) forming, in a medium containing a sample, a complex between the analyte
and at least one antibody specific for the analyte;
(b) analyzing the medium to detect the amount of the complex; and
52

CA 02464472 2004-04-20
WO 03/059251
PCT/US02/33991
(c) relating the amount of the complex to the amount of analyte in the sample.

Such methods may also include forming the complex with at least one antibody
that is
specific for the analyte. The specificity of the antibody is provided by a non-
antibody
targeting agent specific for the analyte which is covalently linked to a
reactive amino
acid in the combining site of the antibody. Thus, the antibody targeting
compounds of
the invention can be used in immunoassays for detecting and measuring the
amount of
an analyte in a sample as has been done previously with conventionally
prepared
polyclonal or monoclonal antibodies. Such assays are well known in the art and

include RIA, ETA, Western, ELISA, and the like. The assay formats may be
competitive or non-competitive and may be direct or indirect. The antibody
targeting
compound can be used in the liquid phase and/or can be bound to a solid phase
carrier.
Carriers include glass, polystyrene, polypropylene, polyethylene, dextran,
nylon,
natural and modified cellulose, polyacrylamide, agarose, magnetite, and the
like. The
nature of the carrier can be either soluble or insoluble. The antibody
targeting
compound may be detectably labeled in any of various ways well known in the
art.
U.S. Patent Nos. 4,659,678; 4,780,423; and 4,298,685 are exemplary of such
assays.
[00134] Viewed in general terms, the amount of an analyte in a sample can
be
determined by forming, in a medium containing the sample, a complex between
the
analyte and at least one antibody specific for the analyte. The medium is then
analyzed
to determine the amount of the complex that is formed. Finally, the amount of
complex
formed is then related to the amount of analyte in the sample. As already
described,
this general approach can take many forms such as direct and indirect,
homogenous or
heterogeneous, and competitive and noncompetitive. In all cases, the antibody
targeting compounds of the invention may be used to replace functions provided
by
conventionally prepared antibodies.
[00135] Also provided is a direct or indirect binding assay where the
presence of an
analyte is determined using an antibody specific for the analyte. In such
methods, the
presence of the analyte is determined using an antibody specific for the
analyte. The
antibody specificity results from a non-antibody targeting agent that is
specific for the
analyte, and the targeting agent is covalently linked to a reactive amino acid
in the
53

CA 02464472 2004-04-20
WO 03/059251
PCT/US02/33991
combining site of the antibody. Thus, antibody-targeting compounds of the
invention
can be used in qualitative assays in place of conventionally prepared
antibodies.
[00136] It would be readily evident that the compounds of the invention
find use not
only in human medical therapy and diagnosis but also in veterinary,
agricultural,
= environmental and other disciplines.
[00137] Also provided are methods of inhibiting or reducing the ability of
a
targeting agent or biological agent to cross a cell membrane. In these methods
an
antibody targeting compound is formed by covalently linking the combining site
of an
antibody that does not itself cross the cell membrane to the targeting agent
or biological
agent, wherein linkage of said antibody to said targeting agent or biological
agent
reduces or inhibits the ability of the agent to cross the cell membrane.
Antibodies that
are not directed to cell surface internalizing receptors are a preferred
source of
antibodies that do not cross cell membranes.
[00138] Further provided are methods of mediating intracellular delivery of
a
intracellularly active drug. In these methods, an antibody targeting compound
is
prepared wherein said compound includes one or more targeting agents or one or
more
biological agents or both covalently linked via a linker to the combining site
of the
antibody. The targeting agents or biological agents are characterized in that
they bind
to a cell receptor and mediate internalization of the agent. The antibody
targeting
compound also includes a drug that is active intracellularly. Intracellular
drug delivery
occurs when a cell expressing the receptor contacts the antibody targeting
compound.
The contacting results in internalization of the antibody targeting agent and
delivery of
said drug intracellularly.
[00139] This approach uses takes advantage of receptor mediated endocytosis
(i.e.,
receptor mediated internalization) to deliver the antibody targeting compound
intracellularly. Cell surface receptors that mediate internalization of
binding ligands are
well known in the art and include, for example, integrins, HER2, EGF receptor,
folic
54

CA 02464472 2004-04-20
WO 03/059251
PCT/US02/33991
acid receptor, and the like. Internalization assays are readily available and
can be
evaluated using fluorescent detection methods.
[00140] In some embodiments, the intracellularly active drug is a prodrug
that
becomes active when said drug contacts an intracellular compartment. The
antibody
targeting compound may include an intracellular trafficking signal to direct
the
internalized antibody targeting compound to a particular intracellular
compartment.
Many proteins contain one or more targeting sequences that serve as a
trafficking signal
or address to target the protein to the correct intracellular site. Receptors
at the
destination also may be involved in the trafficking process.
[00141] The sequences that direct proteins and other compounds to different
intracellular sites such as endoplasmic reticulum, endosome, golgi, or
nucleus, and the
like, are well known in the art. For example, endoplasmic reticulum
trafficking signals
include a KDEL or KKXX sequence, golgi trafficking signals include a GRIP
domain
(see Munro et al., Curr Biol 9: 377-379, 1999), lysosomal trafficking signals
(from
golgi) include mannose-6-phosphate modified oligosaccharides, and nuclear
localization trafficking signals which include one or two short positively
charged
sequences, e.g., lysine or arginine rich (see, Penco et al. Biotech Appl
Biochem 34:151-
159 2001).
[00142] The versatility of the invention is illustrated by the following
Examples
which illustrate preferred embodiments of the invention and are not limiting
of the
claims or specification in any way.
EXAMPLE 1: Antibody targeting compound comprising an RGD
peptidomimetic targeting agent covalently linked to the
combining site of aldolase monoclonal antibody 38C2.
[00143] An integrin targeting compound was formed based on the formation of a
reversible covalent bond between a diketone linker derivative of an RGD
peptidomimetic and the reactive lysine of mouse mAb 38C2. Mouse mAb 38C2 is
the

CA 02464472 2004-04-20
WO 03/059251
PCT/US02/33991
prototype for a new class of catalytic antibodies generated by reactive
immunization
and mechanistically mimic natural aldolase enzymes (Barbas et al., Science
278, 2085-
2092, 1997). Through a reactive lysine, these antibodies catalyze aldol and
retro-aldol
reactions using the enamine mechanism of natural aldolases (Wagner et al.,
Science
270, 1797-1800, 1995; Barbas et al., Science 278, 2085-2092, 1997; Zhong et
al.,
Angew. Chem. Int. Ed. 38, 3738-3741, 1999). In addition to their versatility
and
efficacy in synthetic organic chemistry, aldolase antibodies have been used in
the
activation of camptothecin, doxorubicin, and etopo side prodrugs in vitro and
in vivo as
an anti-cancer strategy (Shabat et al., Proc. Natl. Acad. Sci. US.A. 96, 6925-
6930,
1999); Shabat, D. et al. Proc. Natl. Acad. Sci. US.A. 98, 7528-7533, 2001).
Yet
another feature of these antibodies, namely their ability to bind diketones
covalently,
has remained largely unexplored.
[00144] The RGD peptidomimetic used (see Compound 1) is specific for human
integrin with a high binding affinity for avP3 at 0.9 nM and av135 at 0.6 nM
(specificity
exhibited by minimal anbb3 binding) (Miller et al., supra). A diketone linker
modified
version of Compound 1, designated SCS-873, was prepared as described above.
[00145] A peptidomimetic RGD antagonist with known activity for both a.,133 or

avI35 binding is desirable because some of these compounds bind both murine
and
human integrins. Such species cross reactivity affords preclinical in vivo
studies in
animal angiogenesis models prior to human trials. In addition, the targeting
compound
may be used for the therapy of Kaposi's sarcoma which is associated with avP3
integrin.
[00146] SCS-873 was linked to antibody 38C2 by the following procedure: One
milliliter antibody 38C2 in phosphate buffered saline (10mg/m1) was added to
12
microliters of a 10 mg/mL stock solution of SCS-873 and the resulting mixture
was
maintained at room temperature for 2 hours prior to use.
56

CA 02464472 2004-04-20
WO 03/059251
PCT/US02/33991
[00147] The binding of a mixture of SCS-873 and 38C2 to SLK cells was
evaluated.
SCS-873 effectively mediated cell surface binding of 38C2. No binding of 38C2
was
detectable in the absence of SCS-873. Control experiments confirmed that the
diketone
moiety of the linker is required for binding of SCS-873 to 38C2. It was
determined that
SCS-873 retains the integrin specificity of the integrin targeting component,
i.e., no
binding to aubb3 in ELISA was detected while binding to av[33 and av133 was
found to be
strong. Independent i.p. and i.v. injections of the targeting compound
prepared with
SCS-873 and 38C2 versus each component alone into mice demonstrated integrin
targeting in vivo. In these experiments, the serum half-life of SCS-873 was
extended by
more than two orders of magnitude through binding to 38C2. Free SCS-873 not
bound
to antibody had a serum half-life of only minutes while the combination of
antibody
and small molecule could be detected in the serum sampled from eye bleeds
after
several days.
EXAMPLE 2: Antibody targeting compound comprising IL-4 as targeting
agent covalently linked to the combining site of aldolase
monoclonal antibody 38C2.
[00148] Kaposi's sarcoma tumor cells, among other human epithelial tumor
cells,
express interleukin-4 (IL-4) receptors that can be targeted with a recombinant
chimeric
protein consisting of IL-4 and a truncated form of bacterial toxin called
Pseudomonas
exotoxin (Husain et al., 1999, Nat. Med. 5, 817-822). Based on these studies,
an IL-4
targeting compound for targeting mAb 38C2 to Kaposi's sarcoma tumor cells is
prepared. A linker with a diketone reactive group is conjugated to a lysine
side chain of
IL-2 using a lysine reactive moiety such as N-hydroxysuccinimide (NHS).
Alternatively, a recombinant IL-4 with an added free cysteine is used for
conjugation to
cysteine reactive moieties such as maleimide. To reduce immunogenicity
associated
with the linker portion of the targeting compound, the spacer (i.e. linker
connecting
chain) between the diketone reactive group on one end and the NHS or maleimide

group on the other, is a polyethylene glycol (PEG) chain. Long chain PEG and
other
polymers are known for their ability to mask foreign epitopes, resulting in
the reduced
immunogenicity of therapeutic proteins that display foreign epitopes (Katre et
al., 1990,
57

CA 02464472 2004-04-20
WO 03/059251
PCT/US02/33991
Immunol. 144, 209-213; Francis et al., 1998, Int. J. Hematol. 68, 1-18). Not
more
than one to two diketones should be conjugated to the IL-4 in order to avoid
clearance
of cross-linked antibodies (Rehlaender and Cho, 1998, Pharm. Res. 15, 1652-
1656).
Other interleukins such as IL-2 can be used in place of IL-4 as the targeting
agent.
While IL-4 can be used primarily as a targeting module, an enhancement of its
pharmacological effect (Lussow et al., 1996, Transplantation 62, 1703-1708)
may
result from IL-2 receptor triggering due to the prolonged serum half-life of
the
interleukin obtained through its linkage to an antibody.
EXAMPLE 3: Antibody targeting compound comprising VEGF-R2 binding
peptide as targeting agent covalentiv linked to the combining
site of aldolase monoclonal antibody 38C2.
[00149] Vascular endothelial growth factor (VEGF) is a key modulator of
tumor
angiogenesis. Induced by hypoxia, VEGF expression is upregulated through the
induction of VEGF mRNA transcription in the tumor. Following production and
release by the tumor, VEGF diffuses to endothelial cells of nearby preexisting
blood
vessels, which display VEGF receptors (VEGFR). VEGF binds to two tyrosine
kinase
receptors, VEGFR-1 and VEGFR-2, which are expressed predominantly on
endothelial
cells. Activation of endothelial cells is associated with the binding of VEGF
to
VEGFR-2, whereas VEGFR-1 probably functions as a decoy receptor that regulates
the
local concentration of VEGF (Neufeld et al., 1999, FASEB J. 13, 9-22).
Following
activation, the endothelial cells proliferate, migrate directionally toward
the tumor, and
eventually roll up and interconnect to form new blood vessels. Anti-angiogenic
drugs
that interfere with the interaction of VEGF and VEGR-2 are promising
candidates for
cancer therapy (Klohs and Hamby, 1999, Curr. Opin. Biotechnol. 10, 544-549).
Binetruy-Tournaire et al. (2000, EMBO 1 19, 1525-1533) identified the VEGFR-2
binding linear peptide ATWLPPR (SEQ ID NO: 2) through phage display of peptide

libraries. ATWLPPR (SEQ ID NO: 2) effectively interfered with VEGF binding to
VEGFR-2 and inhibited VEGF-mediated angiogenesis.
58

CA 02464472 2004-04-20
WO 03/059251
PCT/US02/33991
[00150] An antibody targeting compound comprising VEGF-R2 binding peptide
is
prepared by synthesizing the peptide with an additional Cys residue at the
amino or
carboxy terminus, resulting in a peptide with the sequence ATWLPPRC (SEQ ID
NO:
3) and CAT WLPPR (SEQ ID NO: 4), respectively. These thiol-modified peptides
are
reacted with a maleimide/diketone linker (FIG. 14) to produce peptide-linker-
diketo
and diketo-linker-peptide. Incubation of these diketone derivatives with
mAb38C2
results in a covalent linkage between the VEGFR-2 peptide and the antibody
combining
site. The resulting antibody - VEGFR-2 targeting compound is used to target
endothelial cells that express VEGFR-2 such as in tumor angiogenesis. The
compound
prolongs the half-life of the peptide and equips it with antibody effector
function.
EXAMPLE 4: Antibody targeting compound comprising neutralizing RNA
aptamer as targeting agent covalently linked to the combining
site of aldolase monoclonal antibody 38C2.
[00151] Using the process of SELEX (Systematic Evolution of Ligands by
Exponential Enrichment), RNA and DNA aptamers to a variety of molecular
targets
have been generated (Jayasena, 1999, Clin. Chem. 45, 1628-1650). For example,
2'
fluoropyrimidine RNA aptamers that include about 25 nucleotides and that bind
VEGF
with an affinity in the 100-pM range were described (Rucicman et at., 1999, 1
Biol.
Chem. 32, 20556-20567). Like the peptide described in the previous example,
the
aptamers were found to interfere with the interaction of VEGF and VEGFR-2.
[00152] An antibody targeting compound comprising VEGF RNA aptamer is
prepared using commercially available thiol-derivatized nucleotides such as 5'-

phosphorothioate. A phosphorothioate group is a modified phosphate group with
one
of the oxygen atoms replaced by a sulfur atom. The thiol-modified nucleotide
within
the RNA aptamer is reacted with a maleimide diketone (e.g., FIG. 14) to
produce an
RNA aptamer targeting-diketone linker compound. Alternatively, a primary amino

group is introduced into the RNA aptamer using commercially available amino
modifiers. A nucleotide labeled with a primary amino group within the RNA
aptamer
is reacted with a linker that has N-hydroxysuccinimide diketone as the
reactive group.
59

CA 02464472 2004-04-20
WO 03/059251
PCT/US02/33991
Incubation of the diketone derivatives with mAb38C2 results in a covalent
linkage
between the RNA aptamer and the antibody combining site. The resulting
antibody ¨
RNA aptamer VEGFR-2 targeting compound is used to target endothelial cells
that
express VEGFR-2 such as in tumor angiogenesis. The compound prolongs the half-
life
of the RNA aptamer and equips it with antibody effector function.
EXAMPLE 5: Antibody targeting compound comprising folate as targeting
agent covalently linked to the combining site of aldolase
monoclonal antibody 38C2.
[00153] The folate receptor mediates the uptake of folic acid into cells by
endocytosis. It is overexpressed on a variety of epithelial tumor cells
(Leamon and
Low, 2001, Drug Discov. Today 6, 44-51). For example, greater than 90 % of
ovarian
carcinomas express the folate receptor (Sudimack and Lee, 2000, Adv. Drug
Deliv. Rev.
41, 147-162). Mabs directed to the folate receptor, for example Mov18 and
Mov19,
have been evaluated as drugs for ovarian cancer therapy (Coney et al., 1994,
Cancer
Res. 54, 2448-2455; Molthoff et al., 1997, Cancer 80, 2712-2720). Folate-
mediated
targeting of cancer cells over expressing the folate receptor is an
alternative strategy
(Leamon and Low, 2001, Drug Discov. Today 6, 44-51). For example,
chemotherapeutic drugs such as maytansinoids (Ladino et al., 1997, Int. I
Cancer 73,
859-864), are conjugated to folate for selective chemotherapy.
[00154] A targeting agent-linker compound comprising folate derivatized
with a
diketone shown in FIG. 2E is linked to mAb 38C2 and is used to target ovarian
cancer
cells. Because a majority of ovarian tumor cells also express integrins avr33
and/or
avi35, in addition to the folate receptor, a dual targeting compound may be
used for
treatment. A targeting agent-linker compound comprising folate and an RGD
peptidomimetic antagonist are together derivatized with a single diketone
linker to form
the dual targeting compound shown in FIG. 4B. The targeting agent-linker is
linked to
mAb 38C2 and is used to target ovarian cancer cells.

CA 02464472 2004-04-20
WO 03/059251
PCT/US02/33991
EXAMPLE 6: Antibody targeting compound comprising an inhibitor of
prostatic acid phosphatase or prostate-specific antigen as
targeting agent covalently linked to the combining site of
aldolase monoclonal antibody 38C2.
[00155] Prostatic acid phosphatase (PAP) and prostate-specific antigen
(PSA), a
serine protease, are expressed on the cell surface of prostate tumor cells and
are used as
markers for prostate cancer. Mabs directed to PAP and PSA have long been
considered
promising drugs for prostate cancer therapy (Chang et al., 1999, Curr. Opin.
Urol. 9,
391-395). More recently, small synthetic molecules that are specific
inhibitors of PAP
(Beers et al., 1996, Bioorg. Med. Chem. 4, 1693-1701) and PSA (Adlington et
al., 2001,
Med. Chem. 44, 1491-1508) have been reported. Other cell surface enzymes
specific
for prostate tumor cells, such as the recently identified serine protease
hepsin (Magee et
al., 2001, Cancer Res. 61, 5692-5696), also can be used as a target after
specific small
synthetic molecules or peptides targeting agents are identified.
[00156] A targeting agent-linker compound comprising a PAP and/or PSA
inhibitor
is derivatized with a diketone linker to form the compound shown in FIG. 2C).
The
targeting agent-linker is linked to mAb 38C2 and is used to target prostate
cancer.
EXAMPLE 7: Antibody targeting compound comprising thrombopoietin
mimetic peptides or small-molecule agonists of the
thrombopoietin receptor covalently linked to the combining
site of aldolase monoclonal antibody 38C2.
[00157] The cell surface thrombopoietin receptor (cMpl, TPOR) is a member
of the
hematopoietic growth factor receptor superfamily. Thrombopoietin (TPO), the
cytokine that binds to the thrombopoietin receptor, plays a central role in
megakaryopoiesis and platelet production. Therapeutically, recombinant TPO is
being
tested in the clinic for the treatment of thrombocytopenia resulting from
chemotherapy
and bone marrow transplantation. As a therapeutic compound, TPO suffers from a
61

CA 02464472 2004-04-20
WO 03/059251
PCT/US02/33991
relatively short half-life in vivo and from manufacturing and formulation
short-
comings.
[00158] A TPO targeting agent antibody compound is prepared to treat
treatment of
thrombocytopenia resulting from chemotherapy and bone marrow transplantation.
The
TPO mimetic peptide AF12505 with the sequence IEGPTLRQWLAARA (SEQ ID NO:
5), which has been reported to mimic the activity of recombinant TPO (Cwirla
et al.,
1997, Science, 276:1696-9), is synthesized with an additional Cys residue
added to the
amino terminus to produce CIEGPTLRQWLAARA (SEQ ED NO: 6). This thiol-
labeled peptide is then reacted with a maleimide/diketone linker (FIG. 14) to
produce
TPO peptide-linker (diketone) compound. Incubation of this diketone derivative
with
mAb38C2 generates an antibody-TPO receptor targeting compound.
[00159] In vitro assays are used to demonstrate that the targeted antibody
binds live
cells expressing the TPOR and stimulated megakaryocyte colony formation to a
greater
extent than the peptide AF12505. Other TPO mimetic peptides are known in the
art
and can also be used as the TPO receptor targeting agent. In addition, small-
molecule
mimetics with TPO receptor binding have recently been described by Kimura et.
al
(FEBS Lett, 1998, :428(3):250-4.) also may be used in preparing TPOR targeting

compounds.
[00160] The above approach can be similarly applied to target the
erythropoietin
(EPO) receptor using EPO targeting mimetics that have increased therapeutic
efficacy
(Middleton et al., J Biol Chem., 1999, 274(20):14163-9; Johnson et al.,
Nephrol Dial
Transplant., 2000, 15(9):1274-7).
EXAMPLE 8: Antibody targeting compound comprising T-20 peptide or
small-molecules that bind the envelope proteins of HIV-1
covalently linked to the combining site of aldolase
monoclonal antibody 38C2.
62

CA 02464472 2004-04-20
WO 03/059251
PCT/US02/33991
[00161] T-20, N-Acetyl-YTSLIFISLIEESQNQQEKNEQELLELDKWASLWNWF
(SEQ JD NO: 7), a synthetic peptide corresponding to a region of the
transmembrane
subunit of the 11W-1 envelope protein, blocks cell fusion and viral entry at
concentrations of less than 2 ng/ml in vitro. When administered intravenously,
T-20
(monotherapy), the peptide decreases plasma HIV RNA levels demonstrating that
viral
entry can be successfully blocked in vivo. Administration of 1-20 provides
potent
inhibition of 11W replication comparable to anti-retroviral regimens approved
at present
(Kilby et al., Nat Med., 1998, 4(11):1302-7). This peptide drug suffers from a
short
half-life in vivo of approximately 2 hrs.
[00162] An antibody targeting compound using the T-20 peptide as targeting
agent
was produced to increase the valency, potency, and half-life of T-20. The T-20
peptide
was synthesized with an additional Cys residue at the carboxy terminus, the
resulting
modified T-20 peptide having the sequence N-Acetyl-YTSLIHSLIEESQNQQEKNE
QELLELDKWASLWNWFC (SEQ ID NO: 8). This thiol-labeled peptide was then
reacted with a maleimide/diketone linker (FIG. 14) to produce a T-20-Cys-
linker
compound. Incubation of this targeting agent-diketone linker with Ab38C2
resulted in
a covalent linkage between the peptide and the antibody. In vitro assays
demonstrated
that the targeted antibody demonstrated increased potency in inhibiting HIV-1
entry
and infection.
[00163] In addition to peptides that target the envelope proteins ofHW-1, a
number
of small-molecules that bind the envelope proteins have been described. For
example,
the betulinic acid derivative IC9564 is a potent anti-human immunodeficiency
virus
(anti-HIV) compound that can inhibit both HIV primary isolates and laboratory-
adapted
strains. Evidence suggests that 11IV-1 gp120 plays a key role in the anti-HIV-
1 activity
of IC9564 (Holz-Smith et al., Antimicrob Agents Chemother., 2001, 45(1):60-6.)

Preparing an antibody targeting compound in which IC9564 is the targeting
agent is
expected to have increased activity over IC9564 itself by increasing valency,
half-life,
and by directing immune killing of HIV-1 infected cells based on the constant
region of
the antibody chosen. Similarly, recent X-ray crystallographic determination of
the
63

CA 02464472 2004-04-20
WO 03/059251
PCT/US02/33991
HIV-1 envelope glycoprotein gp41 core structure opened up a new avenue to
discover
antiviral agents for chemotherapy of HIV-1 infection and AIDS. Compounds with
the
best fit for docking into the hydrophobic cavity within the gp41 core and with

maximum possible interactions with the target site can also be improved by
addition of
a diketone arm and covalent linkage to an antibody. Several compounds of this
class
have been identified (Debnath et al., J Med Chem., 1999, 42(17):3203-9).
EXAMPLE 9: Antibody Targeting compound formation in vivo via
Transgenic expression of the antibody and administration of
the targeting agent-linker derivative.
[00164] Within the scope of the methods of the present invention is in vivo
formation of the targeting compounds of the invention. In one approach, mAb
38C2 is
produced in vivo from an inducible transgene and a targeting agent-linker
derivative
(e.g. diketone linker) is administered. Using gene delivery vectors, such as
adenoviruses, cDNAs encoding light and heavy chain or a single-chain fragment
of
mAb 38C2 can be introduced into a host organism to establish the antibody
transgene.
This approach allows increased flexibility in treatment. For example, a
patient with a
general risk of cancer chooses to receive the transgene prior to the actual
detection of
the disease. Once cancer is diagnosed, expression of the reactive antibody
(e.g. mAb
38C2) is induced in the patient and a targeting agent ¨linker derivative (e.g.
diketone
linker), where the targeting agent is specifically designed for targeting and
affecting the
diagnosed cancer, is administered. Ideally, both transgene induction and drug
administration are accomplished orally, thus avoiding hospitalization.
EXAMPLE 10: Antibody targeting compound libraries with improved
detectability.
[00165] Screening of small molecule or peptide antagonist, agonists, or
simple
binding molecules is often hampered by the assay available for the detection
of the
binding event. Often, displacement or competition assays are required where
the small
molecule displaces or competes with the binding of another molecule to the
target site.
64

CA 02464472 2004-04-20
WO 03/059251
PCT/US02/33991
The assay must frequently be specifically designed for the specific target
molecule.
The direct detection of a small molecule binding to either a cell surface or a
protein is
often not possible.
[00166] This problem is addressed by preparing the library in the form of
antibody
targeting compounds. To this end, small molecule libraries are synthesized
with an
appended reactive group such as a diketone or a high affinity tag such as
biotin.
Incubation of the tagged molecule with the target allows simple and sensitive
detection
of the binding event, accomplished using an enzyme-linked or fluorophore
labeled
antibody (e.g. 38C2 for the diketone) or streptavidin (for biotin). These
types of assays
are readily adapted for high throughput screening of compound and peptide
libraries.
The advantage of this direct screening of tagged molecules is that the
detection method
is sensitive and standardized over the diversity of possible cell surface
molecules and
protein or other soluble protein targets. Once identified, the attachment site
of the
linker arm does not need to be designed since it pre-exists in the tagged
molecule.
Therefore direct addition of the covalent binding antibody provides the novel
therapeutic agent. In cases where a biotin tag is used for detection, the
biotin arm is
readily exchanged for a diketone arm for direct addition of the covalent
binding
antibody providing the novel therapeutic agent. If the library is a biological
display
library such as a peptide phage library, the site of attachment of the
diketone arm is at
the point where the peptide library resides are joined to the phage coat
protein.
EXAMPLE 11: Antibody targeting compound comprising TAK-779 small-
molecules that bind the envelope proteins of HIV-lcovalently
linked to the combining site of aldolase monoclonal antibody
38C2.
[00167] The 13-chemokine receptor CCR5 is an attractive target for
inhibition of
macrophage-tropic (CCR5-using or RS) HIV-1 replication because individuals
having a
nonfunctional receptor (a homozygous 32-bp deletion in the CCR5 coding region)
are
apparently normal, but are resistant to infection with R5 HIV-1. TAK-779 is a
low
molecular weight (Mr 531.13) nonpeptide CCR5-antagonist (Baba et al., (1999,
Proc.

CA 02464472 2004-04-20
WO 03/059251 PCT/US02/33991
Natl. Acad. Sci. USA, 96, 5698-5703). A targeting agent-linker compound was
prepared by derivatizing TAK-779 with a diketo linker to yield the compound
shown in
FIG. 2D. The diketo-TAK-779 compound was incubated with Mab 38C2 to generate
an antibody CCR5 targeting compound (TAK-799 based). This compound displayed
highly potent and selective inhibition of R5 HIV-1 replication and bound
specifically to
CCR5 expressing cells. The antibody CCR5 targeting compound also displayed
increased valency, increased biological potency, and increased serum half-life
over that .
of the TAK-799 itself.
[00168] Other CCR5 antagonists (Shiraishi, et al., 2000, J. Med. Chem., 43,
2049-
2063) can also be modified for reaction with covalent binding antibodies to
produce
targeting compounds of the invention. A wide variety of chemokine receptor
antagonists may also be modified using this approach.
EXAMPLE 12: Antibody targeting compound comprising LHRH peptide
covalentiv linked to the combining site of aldolase
monoclonal antibody 38C2.
[00169] [D-Lys6] LH-RH antagonist Glp-His-Trp-Ser-Tyr-D-Lys-Leu_Arg-Pro-
Gly-NH2 (SEQ ID NO: 9) (100 micromoles) was dissolved in 1 mL anhydrous DMF.
One equivalent of NHS-diketone linker (compound 35) was added with stirring
overnight. Solvent was evaporated in vacuo, and the product was purified by
HPLC.
The resulting [D-Lys6] LH-RH- diketone linker compound was used directly for
coupling to antibody 38C2. The resulting covalently-modified antibody
specifically
bound the OV-1063 human epithelial ovarian cancer line known to express the LH-
RH
receptor.
66

CA 02464472 2012-03-06
28395-106
0 0
0
0 0
N
0
0
36
[00170] The invention thus has been disclosed broadly and illustrated
in
reference to representative embodiments described above. Those skilled in the
art
will recognize that various modifications can be made without departing from
the
scope of the invention as defined by the claims. Definitions that are
contained in text
incorporated by reference are excluded to the extent that they contradict
definitions in
this disclosure. All structures shown herein are contemplated to provide all
enantiomers.
67

CA 02464472 2005-03-04
SEQUENCE LISTING
<110> Barbas, Carlos F, III
Lerner, Richard A
Sinha, Subhash C
Rader, Christoph
<120> Antibody Targeting Compounds
<130> TSRI 789.2/CPI 0005P
<140> US 10/278,364
<141> 2002-10-22
<150> US 60/344,614
<151> 2001-10-22
<150> US 60/412,455
<151> 2002-09-19
<160> 9
<170> FastSEQ for Windows Version 4.0
<210> 1
<211> 5
<212> PRT
<213> Artificial Sequence
<220>
<223> antibody-heavy chain-complementarity determining
regions-2;
HCDR2
<400> 1
Leu Glu Trp Ile Gly
1 5
<210> 2
<211> 7
<212> PRT
<213> homo sapiens
<400> 2
Ala Thr Trp Leu Pro Pro Arg
1 5
<210> 3
<211> 8
<212> PRT
<213> Homo sapiens
<400> 3
Ala Thr Trp Leu Pro Pro Arg Cys
1 5
<210> 4
<211> 8
1

CA 02464472 2005-03-04
<212> PRT
<213> Homo Sapiens
<400> 4
Cys Ala Thr Trp Leu Pro Pro Arg
1 5
<210> 5
<211> 14
<212> PRT
<213> Homo Sapiens
<400> 5
Ile Glu Gly Pro Thr Leu Arg Gln Trp Leu Ala Ala Arg Ala
1 5 10
<210> 6
<211> 15
<212> PRT
<213> Homo Sapiens
<400> 6
Cys Ile Glu Gly Pro Thr Leu Arg Gln Trp Leu Ala Ala Arg Ala
1 5 10 15
<210> 7
<211> 36
<212> PRT
<213> Artificial Sequence
<220>
<223> A synthetic peptide corresponding to a region of
the transmembrane subunit of the HIV-1 envelop
protein, blocks
cell fusion and viral entry at concentrations of
less than
2 ng/ml in vitro.
<221> ACETYLATION
<222> 1
<223> N-acetyl tyrosine, the terminal
amino group is acetylated
<400> 7
Tyr Thr Ser Leu Ile His Ser Leu Ile Glu Glu Ser Gln Asn Gln Gln
1 5 10 15
Glu Lys Asn Glu Gln Glu Leu Leu Glu Leu Asp Lys Trp Ala Ser Leu
20 25 30
Trp Asn Trp Phe
<210> 8
<211> 37
<212> PRT
<213> Artificial Sequence
2

CA 02464472 2005-03-04
<220>
<223> A synthetic peptide corresponding to a region of
the transmembrane subunit of the H:V-1 envelop
protein, blocks
cell fusion and viral entry at concentrations of
less than
2 ng/ml in vitro. This one has a Cysteine at the
C-terminus.
<221> ACETYLATION
<222> 1
<223> N-acetyl tyrosine, the terminal amino group of
this peptide has an acetyl group on it
<400> 8
Tyr Thr Ser Leu Ile His Ser Leu Ile Glu Glu Ser Gin Asn Gin Gln
1 5 10 15
Glu Lys Asn Glu Gin Glu Leu Leu Glu Leu Asp Lys Trp Ala Ser Leu
20 25 30
Trp Asn Trp Phe Cys
<210> 9
<211> 10
<212> PRT
<213> Artificial Sequence
<220>
<223> LH-RH antagonist
<221> MOD_RES
<222> 6
<223> The lysine in this position is a D-lysine
<221> AMIDATION
<222> 10
<223> The C-terminus is amidated with an amino group.
<221> MOD_RES
<222> 1
<223> Xaa = pyroglutamic acid
S-(-)-2-pyrrolidone-5-carbonyl residue,
L-pyroglutamic acid residue
<400> 9
Xaa His Trp Ser Tyr Lys Leu Arg Pro Gly
1 5 10
3

Representative Drawing

Sorry, the representative drawing for patent document number 2464472 was not found.

Administrative Status

For a clearer understanding of the status of the application/patent presented on this page, the site Disclaimer , as well as the definitions for Patent , Administrative Status , Maintenance Fee  and Payment History  should be consulted.

Administrative Status

Title Date
Forecasted Issue Date 2014-01-07
(86) PCT Filing Date 2002-10-22
(87) PCT Publication Date 2003-07-24
(85) National Entry 2004-04-20
Examination Requested 2007-06-01
(45) Issued 2014-01-07
Expired 2022-10-24

Abandonment History

There is no abandonment history.

Payment History

Fee Type Anniversary Year Due Date Amount Paid Paid Date
Application Fee $400.00 2004-04-20
Registration of a document - section 124 $100.00 2004-05-28
Registration of a document - section 124 $100.00 2004-05-28
Registration of a document - section 124 $100.00 2004-05-28
Maintenance Fee - Application - New Act 2 2004-10-22 $100.00 2004-09-07
Maintenance Fee - Application - New Act 3 2005-10-24 $100.00 2005-09-06
Maintenance Fee - Application - New Act 4 2006-10-23 $100.00 2006-09-06
Request for Examination $800.00 2007-06-01
Maintenance Fee - Application - New Act 5 2007-10-22 $200.00 2007-09-05
Maintenance Fee - Application - New Act 6 2008-10-22 $200.00 2008-09-05
Maintenance Fee - Application - New Act 7 2009-10-22 $200.00 2009-09-10
Maintenance Fee - Application - New Act 8 2010-10-22 $200.00 2010-09-08
Maintenance Fee - Application - New Act 9 2011-10-24 $200.00 2011-09-08
Maintenance Fee - Application - New Act 10 2012-10-22 $250.00 2012-09-17
Maintenance Fee - Application - New Act 11 2013-10-22 $250.00 2013-09-11
Final Fee $300.00 2013-10-29
Maintenance Fee - Patent - New Act 12 2014-10-22 $250.00 2014-10-01
Maintenance Fee - Patent - New Act 13 2015-10-22 $250.00 2015-09-30
Maintenance Fee - Patent - New Act 14 2016-10-24 $250.00 2016-09-28
Maintenance Fee - Patent - New Act 15 2017-10-23 $450.00 2017-09-27
Maintenance Fee - Patent - New Act 16 2018-10-22 $450.00 2018-09-26
Maintenance Fee - Patent - New Act 17 2019-10-22 $450.00 2019-10-02
Maintenance Fee - Patent - New Act 18 2020-10-22 $450.00 2020-10-02
Maintenance Fee - Patent - New Act 19 2021-10-22 $459.00 2021-09-22
Owners on Record

Note: Records showing the ownership history in alphabetical order.

Current Owners on Record
THE SCRIPPS RESEARCH INSTITUTE
Past Owners on Record
BARBAS, CARLOS F.
LERNER, RICHARD
RADER, CHRISTOPH
SINHA, SUBHASH C.
Past Owners that do not appear in the "Owners on Record" listing will appear in other documentation within the application.
Documents

To view selected files, please enter reCAPTCHA code :



To view images, click a link in the Document Description column. To download the documents, select one or more checkboxes in the first column and then click the "Download Selected in PDF format (Zip Archive)" or the "Download Selected as Single PDF" button.

List of published and non-published patent-specific documents on the CPD .

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.


Document
Description 
Date
(yyyy-mm-dd) 
Number of pages   Size of Image (KB) 
Abstract 2004-04-20 1 59
Claims 2004-04-20 28 871
Drawings 2004-04-20 15 179
Description 2004-04-20 67 3,249
Cover Page 2004-06-25 1 31
Description 2005-03-04 70 3,357
Claims 2010-08-24 5 175
Description 2010-08-24 73 3,450
Description 2012-03-06 72 3,438
Claims 2012-03-06 5 162
Description 2013-01-17 72 3,437
Claims 2013-01-17 5 151
Description 2013-07-16 72 3,436
Claims 2013-07-16 5 150
Cover Page 2013-12-04 1 34
PCT 2004-04-21 4 208
PCT 2004-04-20 2 65
Assignment 2004-04-20 2 89
Prosecution-Amendment 2004-04-20 1 18
Assignment 2004-05-28 7 476
Correspondence 2004-08-19 2 31
Prosecution-Amendment 2005-03-04 4 93
Prosecution-Amendment 2010-03-11 3 100
Prosecution-Amendment 2007-06-01 1 43
Prosecution-Amendment 2011-09-06 3 152
Correspondence 2013-10-29 2 76
Prosecution Correspondence 2010-08-24 14 480
Prosecution-Amendment 2012-03-06 18 780
Prosecution-Amendment 2012-09-20 2 67
Prosecution-Amendment 2013-01-17 11 402
Prosecution-Amendment 2013-04-15 2 48
Prosecution-Amendment 2013-07-16 4 147

Biological Sequence Listings

Choose a BSL submission then click the "Download BSL" button to download the file.

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.

Please note that files with extensions .pep and .seq that were created by CIPO as working files might be incomplete and are not to be considered official communication.

BSL Files

To view selected files, please enter reCAPTCHA code :